1
|
Li X, Kong Z, Cai K, Qi F, Zhu S. Neopterin mediates sleep deprivation-induced microglial activation resulting in neuronal damage by affecting YY1/HDAC1/TOP1/IL-6 signaling. J Adv Res 2024:S2090-1232(24)00301-1. [PMID: 39029901 DOI: 10.1016/j.jare.2024.07.017] [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: 04/26/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024] Open
Abstract
INTRODUCTION Sleep deprivation (SD) is a common disorder in modern society. Hippocampus is an important region of the brain for learning, memory, and emotions. Dysfunction of hippocampus can lead to severe learning and memory disorder, significantly affecting quality of life. SD is accompanied by hippocampal microglia activation and a surge in inflammatory factors, but the precise mechanism remains unclear. Moreover, the ongoing unknown persists regarding how activated microglia in SD lead to neuronal damage. Topoisomerase 1 (TOP1) plays an essential role in the inflammatory process, including the tumor system and viral infection. In this study, we observed a significant elevation in TOP1 levels in the hippocampus of mice subjected to SD. Therefore, we hypothesize that TOP1 may be implicated in SD-induced microglia activation and neuronal damage. OBJECTIVES To investigate the role of TOP1 in SD-induced microglial activation, neuronal damage, and neurobehavioral impairments, and the molecular basis of SD-induced elevated TOP1 levels. METHODS TOP1-specific knockout mice in microglia were used to study the effects of TOP1 on microglial activation and neuronal damage. Transcription factor prediction, RNA interference, ChIP-qPCR, ChIP-seq database analysis, and luciferase reporter assays were performed to explore the molecular mechanisms of YY1 transcriptional activation. Untargeted metabolic profiling was employed to investigate the material basis of YY1 transcriptional activation. RESULTS Knockdown of TOP1 in hippocampal microglia ameliorates SD-induced microglial activation, inflammatory response, and neuronal damage. Mechanistically, TOP1 mediates the release of IL-6 from microglia, which consequently leads to neuronal dysfunction. Moreover, elevated TOP1 due to SD were associated with neopterin, which was attributed to its promotion of elevated levels of H3K27ac in the TOP1 promoter region by disrupting the binding of YY1 and HDAC1. CONCLUSION The present study reveals that TOP1-mediated microglial activation is critical for SD induced hippocampal neuronal damage and behavioral impairments.
Collapse
Affiliation(s)
- Xuan Li
- Lanzhou University Second Hospital, Lanzhou University, 730030 Lanzhou, China
| | - Ziyu Kong
- School of Basic Medicine, Wuhan University, Wuhan 430071, China
| | - Ke Cai
- Lanzhou University Second Hospital, Lanzhou University, 730030 Lanzhou, China
| | - Fujian Qi
- School of Life Sciences, Lanzhou University, 730030 Lanzhou, China
| | - Sen Zhu
- School of Life Sciences, Lanzhou University, 730030 Lanzhou, China; The First Affiliated Hospital of Medical College, Zhejiang University, Zhejiang 310000, China.
| |
Collapse
|
2
|
Qin Y, Zhao Y, Hu X, Chen X, Jiang YP, Jin XJ, Li G, Li ZH, Yang JH, Zhang GL, Cui SY, Zhang YH. Ganoderma lucidum spore extract improves sleep disturbances in a rat model of sporadic Alzheimer's disease. Front Pharmacol 2024; 15:1390294. [PMID: 38720773 PMCID: PMC11076761 DOI: 10.3389/fphar.2024.1390294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction: Ganoderma lucidum (G. lucidum, Lingzhi) has long been listed as a premium tonic that can be used to improve restlessness, insomnia, and forgetfulness. We previously reported that a rat model of sporadic Alzheimer's disease (sAD) that was induced by an intracerebroventricular injection of streptozotocin (ICV-STZ) showed significant learning and cognitive deficits and sleep disturbances. Treatment with a G. lucidum spore extract with the sporoderm removed (RGLS) prevented learning and memory impairments in sAD model rats. Method: The present study was conducted to further elucidate the preventive action of RGLS on sleep disturbances in sAD rats by EEG analysis, immunofluorescence staining, HPLC-MS/MS and Western blot. Results: Treatment with 720 mg/kg RGLS for 14 days significantly improved the reduction of total sleep time, rapid eye movement (REM) sleep time, and non-REM sleep time in sAD rats. The novelty recognition experiment further confirmed that RGLS prevented cognitive impairments in sAD rats. We also found that RGLS inhibited the nuclear factor-κB (NF-κB)/Nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammatory pathway in the medial prefrontal cortex (mPFC) in sAD rats and ameliorated the lower activity of γ-aminobutyric acid (GABA)-ergic neurons in the parabrachial nucleus (PBN). Discussion: These results suggest that inhibiting the neuroinflammatory response in the mPFC may be a mechanism by which RGLS improves cognitive impairment. Additionally, improvements in PBN-GABAergic activity and the suppression of neuroinflammation in the mPFC in sAD rats might be a critical pathway to explain the preventive effects of RGLS on sleep disturbances in sAD.
Collapse
Affiliation(s)
- Yu Qin
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yan Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, China
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Xiao Hu
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
| | - Xi Chen
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yan-Ping Jiang
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Xue-Jun Jin
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Gao Li
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| | - Zhen-Hao Li
- Zhejiang ShouXianGu Pharmaceutical Co., Ltd., Wuyi, Zhejiang, China
| | - Ji-Hong Yang
- Zhejiang ShouXianGu Pharmaceutical Co., Ltd., Wuyi, Zhejiang, China
| | - Guo-Liang Zhang
- Zhejiang ShouXianGu Pharmaceutical Co., Ltd., Wuyi, Zhejiang, China
| | - Su-Ying Cui
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yong-He Zhang
- Department of Pharmacology, School of Basic Medical Science, Peking University, Beijing, China
- Department of Pharmacy, Yanbian University Hospital, Yanji, China
| |
Collapse
|
3
|
Werner JK, Albrecht J, Capaldi VF, Jain S, Sun X, Mukherjee P, Williams SG, Collen J, Diaz-Arrastia R, Manley GT, Krystal AD, Wickwire E. Association of Biomarkers of Neuronal Injury and Inflammation With Insomnia Trajectories After Traumatic Brain Injury: A TRACK-TBI Study. Neurology 2024; 102:e209269. [PMID: 38547447 PMCID: PMC11210587 DOI: 10.1212/wnl.0000000000209269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/05/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Insomnia affects about one-third of patients with traumatic brain injury and is associated with worsened outcomes after injury. We hypothesized that higher levels of plasma neuroinflammation biomarkers at the time of TBI would be associated with worse 12-month insomnia trajectories. METHODS Participants were prospectively enrolled from 18 level-1 trauma centers participating in the Transforming Research and Clinical Knowledge in Traumatic Brain Injury study from February 26, 2014, to August 8, 2018. Plasma glial fibrillary acidic protein (GFAP), high-sensitivity C-reactive protein (hsCRP), S100b, neuron-specific enolase (NSE), and ubiquitin carboxyl-terminal hydrolase-L1 (UCH-L1) were collected on days 1 (D1) and 14 (D14) after TBI. The insomnia severity index was collected at 2 weeks, 3, 6, and 12 months postinjury. Participants were classified into insomnia trajectory classes based on a latent class model. We assessed the association of biomarkers with insomnia trajectories, controlling for medical and psychological comorbidities and demographics. RESULTS Two thousand twenty-two individuals with TBI were studied. Elevations in D1 hsCRP were associated with persistent insomnia (severe, odds ratio [OR] = 1.33 [1.11, 1.59], p = 0.002; mild, OR = 1.10 [1.02, 1.19], p = 0.011). Similarly, D14 hsCRP elevations were associated with persistent insomnia (severe, OR = 1.27 [1.02, 1.59], p = 0.03). Of interest, D1 GFAP was lower in persistent severe insomnia (median [Q1, Q3]: 154 [19, 445] pg/mL) compared with resolving mild (491 [154, 1,423], p < 0.001) and persistent mild (344 [79, 1,287], p < 0.001). D14 GFAP was similarly lower in persistent (11.8 [6.4, 19.4], p = 0.001) and resolving (13.9 [10.3, 20.7], p = 0.011) severe insomnia compared with resolving mild (20.6 [12.4, 39.6]. Accordingly, increases in D1 GFAP were associated with reduced likelihood of having persistent severe (OR = 0.76 [95% CI 0.63-0.92], p = 0.004) and persistent mild (OR = 0.88 [0.81, 0.96], p = 0.003) compared with mild resolving insomnia. No differences were found with other biomarkers. DISCUSSION Elevated plasma hsCRP and, surprisingly, lower GFAP were associated with adverse insomnia trajectories after TBI. Results support future prospective studies to examine their utility in guiding insomnia care after TBI. Further work is needed to explore potential mechanistic connections between GFAP levels and the adverse insomnia trajectories.
Collapse
Affiliation(s)
- J Kent Werner
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Jennifer Albrecht
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Vincent F Capaldi
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Sonia Jain
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Xiaoying Sun
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Pratik Mukherjee
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Scott G Williams
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Jacob Collen
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Ramon Diaz-Arrastia
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Geoffrey T Manley
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Andrew D Krystal
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Emerson Wickwire
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| |
Collapse
|
4
|
Bazaz MR, Asthana A, Dandekar MP. Chitosan revokes controlled-cortical impact generated neurological aberrations in circadian disrupted mice via TLR4-NLRP3 axis. Eur J Pharmacol 2024; 969:176436. [PMID: 38423243 DOI: 10.1016/j.ejphar.2024.176436] [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: 10/15/2023] [Revised: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
The severity of inevitable neurological deficits and long-term psychiatric disorders in the aftermath of traumatic brain injury is influenced by pre-injury biological factors. Herein, we investigated the therapeutic effect of chitosan lactate on neurological and psychiatric aberrations inflicted by circadian disruption (CD) and controlled-cortical impact (CCI) injury in mice. Firstly, CD was developed in mice by altering sporadic day-night cycles for 2 weeks. Then, CCI surgery was performed using a stereotaxic ImpactOne device. Mice subjected to CCI displayed a significant disruption of motor coordination at 1-, 3- and 5-days post-injury (DPI) in the rotarod test. These animals showed anxiety- and depression-like behaviors in the elevated plus maze and forced-swim test at 14 and 15 DPI, respectively. Notably, mice subjected to CD + CCI exhibited severe cognitive impairment in Y-maze and novel object recognition tasks. The compromised neurological, psychiatric, and cognitive functions were mitigated in chitosan-treated mice (1 and 3 mg/mL). Immunohistochemistry and real-time PCR assay results revealed the magnified responses of prima facie biomarkers like glial-fibrillary acidic protein and ionized calcium-binding adaptor molecule 1 in the pericontusional brain region of the CD + CCI group, indicating aggravated inflammation. We also noted the depleted levels of brain-derived neurotrophic factor and augmented expression of toll-like receptor 4 (TLR4)-leucine-rich-containing family pyrin domain-containing 3 (NLRP3) signaling [apoptosis-associated-speck-like protein (ASC), caspase-1, and interleukin 1-β] in the pericontusional area of CD + CCI group. CCI-induced changes in the astrocyte-glia and aggravated immune responses were ameliorated in chitosan-treated mice. These results suggest that the neuroprotective effect of chitosan in CCI-induced brain injury may be mediated by inhibition of the TLR4-NLRP3 axis.
Collapse
Affiliation(s)
- Mohd Rabi Bazaz
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, 500037, India
| | - Amit Asthana
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, 500037, India
| | - Manoj P Dandekar
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, 500037, India.
| |
Collapse
|
5
|
Naeem A, Prakash R, Kumari N, Ali Khan M, Quaiyoom Khan A, Uddin S, Verma S, Ab Robertson A, Boltze J, Shadab Raza S. MCC950 reduces autophagy and improves cognitive function by inhibiting NLRP3-dependent neuroinflammation in a rat model of Alzheimer's disease. Brain Behav Immun 2024; 116:70-84. [PMID: 38040385 DOI: 10.1016/j.bbi.2023.11.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/11/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023] Open
Abstract
Alzheimer's disease (AD) is the seventh most common cause of mortality and one of the major causes of disability and vulnerability in the elderly. AD is characterized by gradual cognitive deterioration, the buildup of misfolded amyloid beta (Aβ) peptide, and the generation of neurofibrillary tangles. Despite enormous scientific progress, there is no effective cure for AD. Thus, exploring new treatment options to stop AD or at least slow down its progress is important. In this study, we investigated the potential therapeutic effects of MCC950 on NLRP3-mediated inflammasome-driven inflammation and autophagy in AD. Rats treated with streptozotocin (STZ) exhibited simultaneous activation of the NLRP3 inflammasome and autophagy, as confirmed by Western blot, immunofluorescence, and co-immunoprecipitation analyses. MCC950, a specific NLRP3 inhibitor, was intraperitoneally administered (50 mg/kg body weight) to rats with AD-like symptoms induced by intracerebroventricular STZ injections (3 mg/kg body weight). MCC950 effectively suppressed STZ-induced cognitive impairment and anxiety by inhibiting NLRP3-dependent neuroinflammation. Moreover, our findings indicate that MCC950 exerts neuroprotective effects by attenuating autophagy in neuronal cells. The inhibiting effects of MCC950 on inflammasome activation and autophagy were reproduced in vitro, provding further mechansistic insights into MCC950 therapeutic action. Our findings suggest that MCC950 impedes the progression of AD and may also improve cognitive function through the mitigation of autophagy and NLRP3 inflammasome inhibition.
Collapse
Affiliation(s)
- Abdul Naeem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | - Ravi Prakash
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | - Neha Kumari
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | | | - Abdul Quaiyoom Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Sandeep Verma
- Department of Chemistry, Indian Institute of Technology, Kanpur, UP 208016, India
| | - Avril Ab Robertson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India; Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India.
| |
Collapse
|
6
|
Huang J, Li W. Molecular crosstalk between circadian clock and NLRP3 inflammasome signaling in Parkinson's disease. Heliyon 2024; 10:e24752. [PMID: 38268831 PMCID: PMC10803942 DOI: 10.1016/j.heliyon.2024.e24752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 12/12/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Research has recently found that both animal models and patients with PD have circadian dysfunction, accompanied by abnormal expression of circadian genes and proteins, which implies that the circadian clock plays a crucial role in PD etiopathogenesis. In addition, a strong relationship between NLRP3 inflammasome signaling and PD has been observed. Meanwhile, the activation of the NLRP3 inflammasome is highly relevant to dysfunctions of the molecular clock. Therefore, alleviating the neuroinflammation caused by NLRP3 inflammasome signaling by adjusting the abnormal molecular clock may be a potential strategy for preventing and treating PD. In this article, we have reviewed the potential or direct relationship between abnormalities of the circadian clock and NLRP3 inflammasome signaling in PD.
Collapse
Affiliation(s)
- Jiahua Huang
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, 201500, Shanghai, China
- Institute of Neurology, Institutes of Integrative Medicine, Fudan University, 201500, Shanghai, China
| | - Wenwei Li
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, 201500, Shanghai, China
- Institute of Neurology, Institutes of Integrative Medicine, Fudan University, 201500, Shanghai, China
| |
Collapse
|
7
|
Shentu Y, Chen M, Wang H, Du X, Zhang W, Xie G, Zhou S, Ding L, Zhu Y, Zhu M, Zhang N, Du C, Ma J, Chen R, Yang J, Fan X, Gong Y, Zhang H, Fan J. Hydrogen sulfide ameliorates lipopolysaccharide-induced anxiety-like behavior by inhibiting checkpoint kinase 1 activation in the hippocampus of mice. Exp Neurol 2024; 371:114586. [PMID: 37898396 DOI: 10.1016/j.expneurol.2023.114586] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
Hydrogen sulfide (H2S), an endogenous gasotransmitter, exhibits the anxiolytic roles through its anti-inflammatory effects, although its underlying mechanisms remain largely elusive. Emerging evidence has documented that cell cycle checkpoint kinase 1 (Chk1)-regulated DNA damage plays an important role in the neurodegenerative diseases; however, there are few relevant reports on the research of Chk1 in neuropsychiatric diseases. Here, we aimed to investigate the regulatory role of H2S on Chk1 in lipopolysaccharide (LPS)-induced anxiety-like behavior focusing on inflammasome activation in the hippocampus. Cystathionine γ-lyase (CSE, a H2S-producing enzyme) knockout (CSE-/-) mice displayed anxiety-like behavior and activation of inflammasome-mediated inflammatory responses, manifesting by the increase levels of interleukin-1β (IL-1β), IL-6, and ionized calcium-binding adaptor molecule-1 (Iba-1, microglia marker) expression in the hippocampus. Importantly, expression of p-Chk1 and γ-H2AX (DNA damage marker) levels were also increased in the hippocampus of CSE-/- mice. LPS treatment decreased the expression of CSE and CBS while increased p-Chk1 and γ-H2AX levels and inflammasome-activated neuroinflammation in the hippocampus of mice. Moreover, p-Chk1 and γ-H2AX protein levels and cellular immunoactivity were significantly increased while CSE and CBS were markedly decreased in cultured BV2 cells followed by LPS treatment. Treatment of mice with GYY4137, a donor of H2S, inhibited LPS-induced increased in p-Chk1 and γ-H2AX levels, mitigated inflammasome activation and inflammatory responses as well as amelioration of anxiety-like behavior. Notably, SB-218078, a selective Chk1 inhibitor treatment attenuated the effect of LPS on inflammasome activation and inflammatory responses and the induction of anxiety-like behavior. Finally, STAT3 knockdown with AAV-STAT3 shRNA alleviated LPS-induced anxiety-like behavior and inhibited inflammasome activation in the hippocampus, and blockade of NLRP3 with MCC950 attenuated neuroinflammation induction and ameliorated LPS-induced anxiety-like behavior. Overall, this study indicates that downregulation of Chk1 activity by H2S activation may be considered as a valid strategy for preventing the progression of LPS-induced anxiety-like behavior.
Collapse
Affiliation(s)
- Yangping Shentu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Mengfan Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hui Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaotong Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Institute of Cixi Biomedical Research, Wenzhou Medical University, Cixi, Zhejiang 315302, China
| | - Wenjing Zhang
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Guizhen Xie
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shaoyan Zhou
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lu Ding
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yun Zhu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Min Zhu
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Nan Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Congkuo Du
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianshe Ma
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ran Chen
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jinge Yang
- Department of Medical Technology, Jiangxi Medical College, Shangrao, Jiangxi 334709, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Institute of Cixi Biomedical Research, Wenzhou Medical University, Cixi, Zhejiang 315302, China.
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Institute of Cixi Biomedical Research, Wenzhou Medical University, Cixi, Zhejiang 315302, China.
| |
Collapse
|
8
|
Zhu M, Huang H. The Underlying Mechanisms of Sleep Deprivation Exacerbating Neuropathic Pain. Nat Sci Sleep 2023; 15:579-591. [PMID: 37533626 PMCID: PMC10392808 DOI: 10.2147/nss.s414174] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Pain disrupts sleep, and sleep deprivation or interference can alter pain perception in animals and humans, for example by increasing sensitivity to pain. However, the mechanism by which sleep affects neuropathic pain remains unclear. In this review, we discuss the available evidence from the epidemiologic, clinical, and human, as well as laboratory studies. In previous studies, we have found that sleep deprivation affects various injurious systems, including opioids, dopaminergic, immune, orexins, hypothalamic-pituitary-adrenal axis, and adenosine. At the same time, these systems play a crucial role in neuropathic pain regulation. In the complex interactions between these neurobiological systems, there may be potential regulatory pathways through which sleep deprivation amplifies neuropathic pain. Because of the impact sleep problems and neuropathic pain can have on the patients' quality of life, studying the link between sleep and neuropathic pain is important for neuropathic pain prevention and public health.
Collapse
Affiliation(s)
- Manmin Zhu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Guizhou, People’s Republic of China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Guizhou, People’s Republic of China
| |
Collapse
|
9
|
Gao C, Cao Y, He M, Zhang X, Zhong Q, Tang L, Chen T, Zhang Z. SAG treatment ameliorates memory impairment related to sleep loss by upregulating synaptic plasticity in adolescent mice. Behav Brain Res 2023; 450:114468. [PMID: 37148913 DOI: 10.1016/j.bbr.2023.114468] [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: 01/28/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 05/08/2023]
Abstract
Adequate sleep during the developmental stage can promote learning and memory functions because synaptic protein synthesis at primed synapses during sleep profoundly affects neurological function. The Sonic hedgehog (Shh) signaling pathway affects neuroplasticity in the hippocampus during the development of the central nervous system. In this study, the changes in synaptic morphology and function induced by sleep deprivation and the potential therapeutic effect of a Shh agonist (SAG) on these changes were investigated in adolescent mice. Adolescent mice were subjected to sleep deprivation for 20 hrs (2pm to 10 am the next day) and were free to sleep for the remaining 4 hrs per day for 10 consecutive days. Sleep-deprived mice were injected with SAG (10mg/kg body weight, i.p.) or saline (i.p.) every day 5min before the onset of the 20h sleep deprivation period. Chronic sleep deprivation impaired recognition and spatial memory, decreased the number of dendritic spines and mEPSCs of hippocampal CA1 pyramidal neurons, decreased the postsynaptic density, and reduced Shh and glioma-associated oncogene homolog 1 (Gli1) expression. SAG significantly protected against sleep deprivation-induced memory dysfunction, increased the CA1 pyramidal neuronal dendritic spine number and mEPSC frequency, and increased Gli1 expression. In conclusion, sleep deprivation induces memory impairment in adolescent mice, and SAG treatment prevents this impairment, probably by enhancing synaptic function in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Chenyi Gao
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yue Cao
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Mengying He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xuemin Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Lijuan Tang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ting Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
10
|
NLRP3 upregulation related to sleep deprivation-induced memory and emotional behavior changes in TRPV1 -/- mice. Behav Brain Res 2023; 440:114255. [PMID: 36563905 DOI: 10.1016/j.bbr.2022.114255] [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: 09/06/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022]
Abstract
Sleep deprivation, which is a common problem in modern society, impairs memory function and emotional behavior. TRPV1, a subfamily of transient receptor potential cation channels, is abundantly expressed in the central nervous system and is associated with animal behavior. In this article, we report that TRPV1 deficiency in mice alleviates sleep deprivation-induced abnormal behaviors. We found that in the sleep-deprived mice, TRPV1 knockout increased the duration and visits in the central area in the open field task and increased visits to the open arms in the elevated plus maze. The TRPV1-/- mice performed better during the test stage in the Morris water maze phase after sleep deprivation. In the mPFC and hippocampus regions, western blotting results showed that TRPV1-/- attenuated sleep deprivation-induced increases in GFAP, NLRP3, and ASC and increased the expression of the mitochondrial marker Tom20. Immunofluorescence results showed that the action of TRPV1 knockout on NLRP3 was negatively correlated with Tom20 after sleep deprivation. Our results confirm that TRPV1 knockout attenuates sleep deprivation-induced behavioral disorders. The effect of TRPV1 on the behavior of sleep-deprived mice may be related to the neuroinflammation associated with mitochondria in the mPFC and hippocampus.
Collapse
|
11
|
Chronic Fatigue, Depression and Anxiety Symptoms in Long COVID Are Strongly Predicted by Neuroimmune and Neuro-Oxidative Pathways Which Are Caused by the Inflammation during Acute Infection. J Clin Med 2023; 12:jcm12020511. [PMID: 36675440 PMCID: PMC9865328 DOI: 10.3390/jcm12020511] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/13/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Long-term coronavirus disease 2019 (long COVID) is associated with physio-somatic (chronic fatigue syndrome and somatic symptoms) and affective (depression and anxiety) symptoms. The severity of the long COVID physio-affective phenome is largely predicted by increased peak body temperature (BT) and lowered oxygen saturation (SpO2) during the acute infectious phase. This study aims to delineate whether the association of BT and SpO2 during the acute phase and the long COVID physio-affective phenome is mediated by neurotoxicity (NT) resulting from activated immune-inflammatory and oxidative stress pathways. METHODS We recruited 86 patients with long COVID (3-4 months after the acute phase) and 39 healthy controls and assessed serum C-reactive protein (CRP), caspase 1, interleukin (IL) 1β, IL-18, IL-10, myeloperoxidase (MPO), advanced oxidation protein products (AOPPs), total antioxidant capacity (TAC), and calcium (Ca), as well as peak BT and SpO2 during the acute phase. RESULTS Cluster analysis revealed that a significant part (34.9%) of long COVID patients (n = 30) show a highly elevated NT index as computed based on IL-1β, IL-18, caspase 1, CRP, MPO, and AOPPs. Partial least squares analysis showed that 61.6% of the variance in the physio-affective phenome of long COVID could be explained by the NT index, lowered Ca, and peak BT/SpO2 in the acute phase and prior vaccinations with AstraZeneca or Pfizer. The most important predictors of the physio-affective phenome are Ca, CRP, IL-1β, AOPPs, and MPO. CONCLUSION The infection-immune-inflammatory core of acute COVID-19 strongly predicts the development of physio-affective symptoms 3-4 months later, and these effects are partly mediated by neuro-immune and neuro-oxidative pathways.
Collapse
|
12
|
Sleep-Disturbance-Induced Microglial Activation Involves CRH-Mediated Galectin 3 and Autophagy Dysregulation. Cells 2022; 12:cells12010160. [PMID: 36611953 PMCID: PMC9818437 DOI: 10.3390/cells12010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Chronic sleep disturbances (CSDs) including insomnia, insufficient sleep time, and poor sleep quality are major public health concerns around the world, especially in developed countries. CSDs are major health risk factors linked to multiple neurodegenerative and neuropsychological diseases. It has been suggested that CSDs could activate microglia (Mg) leading to increased neuroinflammation levels, which ultimately lead to neuronal dysfunction. However, the detailed mechanisms underlying CSD-mediated microglial activation remain mostly unexplored. In this study, we used mice with three-weeks of sleep fragmentation (SF) to explore the underlying pathways responsible for Mg activation. Our results revealed that SF activates Mg in the hippocampus (HP) but not in the striatum and prefrontal cortex (PFc). SF increased the levels of corticotropin-releasing hormone (CRH) in the HP. In vitro mechanism studies revealed that CRH activation of Mg involves galectin 3 (Gal3) upregulation and autophagy dysregulation. CRH could disrupt lysosome membrane integrity resulting in lysosomal cathepsins leakage. CRHR2 blockage mitigated CRH-mediated effects on microglia in vitro. SF mice also show increased Gal3 levels and autophagy dysregulation in the HP compared to controls. Taken together, our results show that SF-mediated hippocampal Mg activation involves CRH mediated galectin 3 and autophagy dysregulation. These findings suggest that targeting the hippocampal CRH system might be a novel therapeutic approach to ameliorate CSD-mediated neuroinflammation and neurodegenerative diseases.
Collapse
|
13
|
Amini M, Yousefi Z, Ghafori SS, Hassanzadeh G. Sleep deprivation and NLRP3 inflammasome: Is there a causal relationship? Front Neurosci 2022; 16:1018628. [PMID: 36620464 PMCID: PMC9815451 DOI: 10.3389/fnins.2022.1018628] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
In the modern era, sleep deprivation (SD) is one of the most common health problems that has a profound influence on an individual's quality of life and overall health. Studies have identified the possibility that lack of sleep can stimulate inflammatory responses. NLRP3 inflammasome, a key component of the innate immune responses, initiates inflammatory responses by enhancing proinflammatory cytokine release and caspase-1-mediated pyroptosis. In this study, NLRP3 modification, its proinflammatory role, and potential targeted therapies were reviewed with regard to SD-induced outcomes. A growing body of evidence has showed the importance of the mechanistic connections between NLRP3 and the detrimental consequences of SD, but there is a need for more clinically relevant data. In animal research, (i) some animals show differential vulnerability to the effects of SD compared to humans. (ii) Additionally, the effects of sleep differ depending on the SD technique employed and the length of SD. Moreover, paying attention to the crosstalk of all the driving factors of NLRP3 inflammasome activation such as inflammatory responses, autonomic control, oxidative stress, and endothelial function is highly recommended. In conclusion, targeting NLRP3 inflammasome or its downstream pathways for therapy could be complicated due to the reciprocal and complex relationship of SD with NLRP3 inflammasome activation. However, additional research is required to support such a causal claim.
Collapse
Affiliation(s)
- Mohammad Amini
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Yousefi
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Sayed Soran Ghafori
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran,*Correspondence: Gholamreza Hassanzadeh,
| |
Collapse
|
14
|
Javanmehr N, Saleki K, Alijanizadeh P, Rezaei N. Microglia dynamics in aging-related neurobehavioral and neuroinflammatory diseases. J Neuroinflammation 2022; 19:273. [PMID: 36397116 PMCID: PMC9669544 DOI: 10.1186/s12974-022-02637-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Microglia represent the first line of immune feedback in the brain. Beyond immune surveillance, they are essential for maintaining brain homeostasis. Recent research has revealed the microglial cells' spatiotemporal heterogeneity based on their local and time-based functions in brain trauma or disease when homeostasis is disrupted. Distinct "microglial signatures" have been recorded in physiological states and brain injuries, with discrete or sometimes overlapping pro- and anti-inflammatory functions. Microglia are involved in the neurological repair processes, such as neurovascular unit restoration and synaptic plasticity, and manage the extent of the damage due to their phenotype switching. The versatility of cellular phenotypes beyond the classical M1/M2 classification, as well as the double-edge actions of microglia in neurodegeneration, indicate the need for further exploration of microglial cell dynamics and their contribution to neurodegenerative processes. This review discusses the homeostatic functions of different microglial subsets focusing on neuropathological conditions. Also, we address the feasibility of targeting microglia as a therapeutic strategy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
15
|
Xie H, Peng J, Zhang X, Deng L, Ding Y, Zuo X, Wang F, Wu Y, Zhang J, Zhu Q. Effects of mitochondrial reactive oxygen species-induced NLRP3 inflammasome activation on trichloroethylene-mediated kidney immune injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114067. [PMID: 36087465 DOI: 10.1016/j.ecoenv.2022.114067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 06/15/2023]
Abstract
This study aimed to investigate the activating mechanism of the NLRP3 inflammasome in trichloroethylene-sensitized mice. In total, 88 BALB/c female mice were used to establish the trichloroethylene (TCE)-sensitized mouse model. Some of the mice received MitoTEMPO, MCC 950 or soluble recombinant CD59-Cys to inhibit mitochondrial reactive oxygen species (mtROS) production, NLRP3 assembly, or C5b-9 formation. Mouse tubular epithelial cell expression levels of NLRP3, ASC, Caspase 1, IL-1β, IL-18 and mitochondrial antiviral signaling protein (MAVS) were detected by western blot. Mitochondrial numbers, membrane potential (ΔΨm) and mtROS were detected by using MitoScene Green II, JC-1 dye and MitoSOX Red indicator, respectively. Tubular epithelial cell calcium levels were detected by a Fluo-8 no wash calcium assay kit. Human kidney-2 (HK-2) cells were cultured and stimulated by C5b6 and normal human serum (NHS) to verify the role of C5b-9-induced mitochondrial ROS in activating NLRP3 inflammasome. Urine α1-MG, β2-MG, and mtROS production and calcium levels were increased, while mitochondrial numbers were decreased in TCE-sensitized positive mice. After treatment with MitoTEMPO, renal tubular injury was alleviated, JC-1 fluorescence and mitochondrial numbers were significantly increased, and mitochondrial ROS were inhibited. The NLRP3 inflammasome was activated in TCE-sensitized positive mice, while Mito TEMPO inhibited MAVS expression and NLRP3 inflammasome activation. The in vitro studies proved that C5b-9 can induce mtROS release and activate the assembly of NLRP3 inflammasome in HK-2 cells. In conclusion, in TCE-sensitized positive mouse renal tubular epithelial cells, C5b-9 caused calcium influx and thus induced mitochondrial injury and mtROS overexpression, finally inducing MAVS expression and NLRP3 inflammasome activation and kidney injury.
Collapse
Affiliation(s)
- Haibo Xie
- Department of Nephropathy, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jiale Peng
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xuesong Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Lihua Deng
- Shenzhen Prevention and Treatment Center for Occupational Disease, Shenzhen, China
| | - Yani Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xulei Zuo
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Feng Wang
- Department of Dermatology, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yonggui Wu
- Department of Nephropathy, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Jiaxiang Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| | - Qixing Zhu
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.
| |
Collapse
|
16
|
de Mello AJ, Moretti M, Rodrigues ALS. SARS-CoV-2 consequences for mental health: Neuroinflammatory pathways linking COVID-19 to anxiety and depression. World J Psychiatry 2022; 12:874-883. [PMID: 36051596 PMCID: PMC9331446 DOI: 10.5498/wjp.v12.i7.874] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/03/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has been linked to an increased prevalence of mental health disorders, particularly anxiety and depression. Moreover, the COVID-19 pandemic has caused stress in people worldwide due to several factors, including fear of infection; social isolation; difficulty in adapting to new routines; lack of coping methods; high exposure to social media, misinformation, and fake reports; economic impact of the measures implemented to slow the contagion and concerns regarding the disease pathogenesis. COVID-19 patients have elevated levels of pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-6, and tumor necrosis factor-α, and other inflammation-related factors. Furthermore, invasion of the central nervous system by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may potentially contribute to neuroinflammatory alterations in infected individuals. Neuroinflammation, a consequence of psychological stress due to the COVID-19 pandemic, may also play a role in the development of anxiety and depressive symptoms in the general population. Considering that neuroinflammation plays a significant role in the pathophysiology of depression and anxiety, this study investigated the effects of SARS-CoV-2 on mental health and focused on the impact of the COVID-19 pandemic on the neuroinflammatory pathways.
Collapse
Affiliation(s)
- Anna Julie de Mello
- Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88040-200, Brazil
| | - Morgana Moretti
- Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88040-200, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88040-200, Brazil
| |
Collapse
|
17
|
Roflumilast, a Phosphodiesterase-4 Inhibitor, Ameliorates Sleep Deprivation-Induced Cognitive Dysfunction in C57BL/6J Mice. ACS Chem Neurosci 2022; 13:1938-1947. [PMID: 35736514 DOI: 10.1021/acschemneuro.2c00127] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Sleep deprivation (SD) interferes with long-term memory and cognitive functions by overactivation of phosphodiesterase (PDEs) enzymes. PDE4, a nonredundant regulator of the cyclic nucleotides (cAMP), is densely expressed in the hippocampus and is involved in learning and memory processes. In the present study, we investigated the effects of Roflumilast (ROF), a PDE4B inhibitor, on sleep deprivation-induced cognitive dysfunction in a mouse model. Memory assessment was performed using a novel object recognition task, and the hippocampal cAMP level was estimated by the ELISA method. The alterations in the expressions of PDE4B, amyloid-β (Aβ), CREB, BDNF, and synaptic proteins (Synapsin I, SAP 97, PSD 95) were assessed to gain insights into the possible mechanisms of action of ROF using the Western blot technique. Results show that ROF reversed SD-induced cognitive decline in mice. ROF downregulated PDE4B and Aβ expressions in the brain. Additionally, ROF improved the cAMP level and the protein expressions of synapsin I, SAP 97, and PSD 95 in the hippocampal region of SD mice. Taken together, these results suggest that ROF can suppress the deleterious effects of SD-induced cognitive dysfunction via the PDE4B-mediated cAMP/CREB/BDNF signaling cascade.
Collapse
|
18
|
Kwon S, Cheon SY. Influence of the inflammasome complex on psychiatric disorders: clinical and preclinical studies. Expert Opin Ther Targets 2021; 25:897-907. [PMID: 34755582 DOI: 10.1080/14728222.2021.2005027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The innate immune complex, an inflammasome complex, has a role in the etiology of psychiatric disorders. Preclinical studies have demonstrated that the inflammasome activation leads to psychiatric disorders and clinical studies have proved that specific psychiatric illnesses are associated with aberrant levels of inflammatory cytokines and inflammasome. The inflammasome complex could be a major factor in the progression and pathology of psychiatric disorders. AREA COVERED We discuss the pathogenesis of psychiatric disorders with respect to the activation of the inflammasome complex. Inflammasome-associated inflammatory cytokines are observed in patients and animal models of psychiatric disorders. The article also reflects on inflammasome regulatory options for the prevention and treatment of psychiatric disorders. Relevant literature available on PubMed from 1992 to 2021 has been included in this review. EXPERT OPINION Modulating the inflammasome complex is a potential therapeutic strategy to treat symptom severity for patients with psychiatric disorders, particularly those with inflammasome-associated disorders. However, the nature of the psychiatric disorders should be considered when targeting inflammasome.
Collapse
Affiliation(s)
- Sunghark Kwon
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea
| | - So Yeong Cheon
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea.,Research Institute for Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea
| |
Collapse
|
19
|
Benedetti F, Dallaspezia S, Melloni EMT, Lorenzi C, Zanardi R, Barbini B, Colombo C. Effective Antidepressant Chronotherapeutics (Sleep Deprivation and Light Therapy) Normalize the IL-1β:IL-1ra Ratio in Bipolar Depression. Front Physiol 2021; 12:740686. [PMID: 34539454 PMCID: PMC8440979 DOI: 10.3389/fphys.2021.740686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022] Open
Abstract
Background Mood disorders associate with peripheral markers of low-grade inflammation, among which circulating levels of interleukin-1β (IL-1β) consistently predict diagnosis and poor outcomes. Antidepressant chronotherapeutics (total sleep deprivation and light therapy, TSD+LT) prompts response in drug-resistant bipolar depression, but its effect on peripheral inflammation were never assessed. Here we explored the effects of TSD+LT on IL-1β signaling. Methods We studied the ratio between IL-1β and its receptor antagonist (IL-1β:IL1ra) in 33 healthy participants, and in 26 inpatients with a major depressive episode in course of Bipolar Disorder, before and after treatment with three cycles of repeated TSD+LT, interspersed with sleep recovery nights, administered during 1 week. Treatment effects of mood and on IL-1β:IL1ra were analyzed in the context of the Generalized Linear Model (GLM). Results At baseline, patients had higher IL-1β, IL1ra, and IL-1β:IL1ra than controls. Treatment significantly decreased IL-1β:IL1ra, by decreasing IL-1β and increasing IL1ra, the effect being proportional to baseline levels and normalizing values. Patients with higher baseline levels showed the highest decrease in IL-1β:IL-1ra, which associated with the immediate antidepressant response at the first cycle; while patients with lower baseline values showed negligible changes in the IL-1β:IL-1ra, unrelated to treatment response. Conclusion We observed a parallel change of inflammatory biomarkers and severity of depression after chronotherapeutics, suggesting that a reduction in inflammation associated with depression could contribute to the mechanism of action of TSD+LT, and warranting interest for controlled studies addressing the role of inflammation in the recovery from bipolar depression.
Collapse
Affiliation(s)
- Francesco Benedetti
- Vita-Salute San Raffaele University, Milan, Italy.,Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | - Sara Dallaspezia
- Vita-Salute San Raffaele University, Milan, Italy.,Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | - Elisa Maria Teresa Melloni
- Vita-Salute San Raffaele University, Milan, Italy.,Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | - Raffaella Zanardi
- Vita-Salute San Raffaele University, Milan, Italy.,Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milan, Italy
| | - Barbara Barbini
- Vita-Salute San Raffaele University, Milan, Italy.,Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milan, Italy
| | - Cristina Colombo
- Vita-Salute San Raffaele University, Milan, Italy.,Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milan, Italy
| |
Collapse
|