1
|
Xiao R, Pan J, Yang M, Liu H, Zhang A, Guo X, Zhou S. Regulating astrocyte phenotype by Lcn2 inhibition toward ischemic stroke therapy. Biomaterials 2025; 317:123102. [PMID: 39836995 DOI: 10.1016/j.biomaterials.2025.123102] [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: 11/20/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
Astrocytes can be reacted to "reactive astrocytes" after ischemia-reperfusion injury, in which A1 phenotype causes neuronal and oligodendrocyte death, whereas the A2 phenotype exerts neuroprotective effects, thus regulating reactive astrocyte to A2 type is a potential target for stroke therapy. Lcn2 level is highly associated with the phenotypic polarization of astrocytes. We found that silencing the Lcn2 gene by adeno-associated virus (AAV)-Lcn2 shRNA adenovirus resulted in a dramatic decrease in A1-type astrocytes and increase in A2 astrocytes in MCAO mice. Hence, a nanoplatform was developed for stroke therapy by inhibiting Lcn2. This system was fabricated by N-acetyl Pro-Gly-Pro peptide-decorated rod-shaped poly (lactic-co-glycolic acid) nanoparticles loading with rolipram (AP@R). The nanodrug can be efficiently taken up by neutrophils simultaneously through morphology-mediated passive targeting and Cxcr2 receptor-mediated active targeting, subsequently crossing the blood-brain barrier (BBB) by hitchhiking neutrophils. When accumulating at the brain parenchyma, the released rolipram can inhibit the Lcn2 level, thereby reversing the astrocyte phenotype to alleviate neuroinflammation and promote BBB repair. This work provides a new strategy for treating ischemic stroke.
Collapse
Affiliation(s)
- Renmin Xiao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Jingmei Pan
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Mengyi Yang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China; Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Hua Liu
- Department of neurology, the third people's hospital of Chengdu & the affiliated hospital of Southwest Jiaotong university, Chengdu 610031, PR China
| | - Aohan Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Xing Guo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China.
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| |
Collapse
|
2
|
Sha R, Chen Y, Xu T, Yang G, Xu L, Xie HQ, Zhao B. Astrocytes Respond to Environment-Relevant Doses TCDD in a Specific Manner Which Is Different from the Endogenous AhR Ligand (FICZ). ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:392-401. [PMID: 40270526 PMCID: PMC12012657 DOI: 10.1021/envhealth.4c00189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 04/25/2025]
Abstract
Astrocytes play an important role in the nervous system's response to external stimulation. Environmental pollutants could activate astrocytes into A1 (toxic) or A2 (protective) types and induce different effects. Meanwhile, the aryl hydrocarbon receptor (AhR) is an environmental molecule sensor in the body and has various ligands. But the difference between exogenous and endogenous AhR ligands on the astrocytic activation is unclear; in this study, we employed rat primary cultured cortical astrocytes to reveal the effects and mechanisms of AhR ligands on astrocytic activation. We found that, after treatment with exogenous AhR ligand (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD) ranging from 0.01 to 0.1 nmol/L, astrocytes mainly exhibited A2 type activation. The specific manifestation includes the increase in the expression of A2 marker genes, the enhancement of cellular autonomous movement, the expression and secretion of chemokines, such as Cxcl10, Cxcl2, and Ccl7. And TCDD-induced A2 type astrocytes show a positive impact on neuronal synaptic formation. Although both TCDD and endogenous AhR ligand (6-formylindolo[3,2-b] carbazole, FICZ) could activate AhR pathway in astrocytes, FICZ (50 nmol/L) neither induces activation of A2 type astrocytes nor upregulation of chemokines. Therefore, our findings suggest that AhR is crucial for astrocytes to recognize environmental pollutants and protect the nervous system.
Collapse
Affiliation(s)
- Rui Sha
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yangsheng Chen
- School
of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Tong Xu
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Guanglei Yang
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Xu
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Heidi Qunhui Xie
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Zhao
- School
of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
3
|
Gao CY, Pan YJ, Su WS, Wu CY, Chang TY, Yang FY. Abdominal ultrasound stimulation alleviates DSS-induced colitis and behavioral disorders in mice by mediating the microbiota-gut-brain axis balance. Neurotherapeutics 2025; 22:e00494. [PMID: 39580323 PMCID: PMC12014354 DOI: 10.1016/j.neurot.2024.e00494] [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: 08/02/2024] [Revised: 10/17/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Inflammatory bowel disease (IBD) has the potential to induce neuroinflammation, which may increase the risk of developing neurodegenerative disorders. Ultrasound stimulation to the abdomen is a potential treatment for dextran sulfate sodium (DSS)-induced acute colitis. The present study aimed to investigate whether abdominal low-intensity pulsed ultrasound (LIPUS) can alleviate DSS-induced neuroinflammation through the microbiota-gut-brain axis. Male mice were fed DSS to induce ulcerative colitis. LIPUS stimulation was then applied to the abdomen at intensities of 0.5 and 1.0 W/cm2. Mouse biological samples were analyzed, and behavior was evaluated. [18F]FEPPA PET/CT imaging was employed to track and quantify inflammation in the abdomen and brain. Changes in the gut microbiota composition were analyzed using 16S rRNA sequencing. Abdominal LIPUS significantly inhibited the DSS-induced inflammatory response, repaired destroyed crypts, and partially preserved the epithelial barrier. [18F]FEPPA accumulation in the colitis-induced neuroinflammation in the abdomen and specific brain regions significantly decreased after LIPUS treatment. LIPUS maintained intestinal integrity by increasing zonula occludens and occludin levels, reduced lipopolysaccharide-binding protein and lipopolysaccharide levels in the serum, and improved behavioral dysfunctions. Moreover, LIPUS, at an intensity of 0.5 W/cm2, reshaped the gut microbiota in colitis-induced mice by increasing the relative abundance of the Firmicutes and decreasing the relative abundance of the Bacteroidota. Our findings demonstrated that abdominal LIPUS stimulation has the potential to be a novel therapeutic strategy to improve colitis-induced behavioral disorders through microbiota-gut-brain axis signaling.
Collapse
Affiliation(s)
- Cong-Yong Gao
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ju Pan
- Department of Psychiatry, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan City, Taiwan
| | - Wei-Shen Su
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Yu Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
4
|
Kim JH, Jeong HG, Hyeon SJ, Park U, Oh WJ, Hwang J, Lim HH, Ko PW, Lee HW, Lee WH, Ryu H, Suk K. Crosstalk between lipocalin-2 and IL-6 in traumatic brain injury: Closely related biomarkers. Exp Neurol 2025; 385:115092. [PMID: 39637963 DOI: 10.1016/j.expneurol.2024.115092] [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: 06/25/2024] [Revised: 11/19/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Clinical biomarkers are crucial for diagnosing and predicting outcomes in patients with traumatic brain injury (TBI). In this study, we performed an unbiased analysis of plasma proteins in acute TBI patients using bead-based multiplex assays and identified a strong positive correlation between LCN2 and IL-6 levels. Based on these findings, we hypothesized that LCN2 and IL-6 are closely related circulating biomarkers for TBI. Our previous and current studies demonstrate that the expression of LCN2, IL-6, and its receptors is upregulated in patients with chronic traumatic encephalopathy, in mouse models of traumatic and ischemic injury, and in an in vitro scratch injury model. Lcn2-deficiency reduced the injury-induced expression of IL-6 and its receptors in both animal and scratch injury models. These results suggest an augmented LCN2-dependent IL-6 signaling in the injured brain. As both LCN2 and IL-6 are secreted proinflammatory mediators, we further explored the possibility of cross-regulation between LCN2 and IL-6. In cultured glial cells, treatment with recombinant LCN2 protein enhanced the microglial expression of IL-6, while IL-6 protein treatment increased astrocytic LCN2 expression. Moreover, IL-6 expression and release were elevated in LCN2-overexpressing transgenic mice. Mechanistically, IL-6 enhanced astrocytic LCN2 expression through STAT3 signaling, while LCN2 upregulated microglial IL-6 expression through the NF-κB pathway. Taken together, our results suggest an important role of the LCN2-IL-6 axis in amplifying neuroinflammation through a positive feedback loop in secondary brain injury conditions. Finally, this study implies the utility of LCN2 and IL-6 as closely related biomarkers for TBI diagnosis and prognosis.
Collapse
Affiliation(s)
- Jae-Hong Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu 41940, Republic of Korea
| | - Han-Gil Jeong
- Division of Neurocritical Care, Department of Neurosurgery and Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si 13620, Republic of Korea
| | - Seung Jae Hyeon
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Uiyeol Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Won-Jong Oh
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41068, Republic of Korea
| | - Junmo Hwang
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41068, Republic of Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41068, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Pan-Woo Ko
- Department of Neurology, Kyungpook National University School of Medicine, Daegu 41404, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Ho-Won Lee
- Department of Neurology, Kyungpook National University School of Medicine, Daegu 41404, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Won-Ha Lee
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea; School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Hoon Ryu
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Veterans Affairs Boston Healthcare System, Boston, MA 02130, United States; Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA 02118, United States.
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu 41940, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
5
|
Thergarajan P, O'Brien TJ, Jones NC, Ali I. Ligand-receptor interactions: A key to understanding microglia and astrocyte roles in epilepsy. Epilepsy Behav 2025; 163:110219. [PMID: 39693861 DOI: 10.1016/j.yebeh.2024.110219] [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: 08/14/2024] [Revised: 11/30/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
Epilepsy continues to pose significant social and economic challenges on a global scale. Existing therapeutic approaches predominantly revolve around neurocentric mechanisms, and fail to control seizures in approximately one-third of patients. This underscores the pressing need for novel and complementary treatment approaches to address this gap. An increasing body of literature points to a role for glial cells, including microglia and astrocytes, in the pathogenesis of epilepsy. Notably, microglial cells, which serve as pivotal inflammatory mediators within the epileptic brain, have received increasing attention over recent years. These immune cells react to epileptogenic insults, regulate neuronal processes, and play diverse roles during the process of epilepsy development. Additionally, astrocytes, another integral non-neuronal brain cells, have garnered increasing recognition for their dynamic contributions to the pathophysiology of epilepsy. Their complex interactions with neurons and other glial cells involve modulating synaptic activity and neuronal excitability, thereby influencing the aberrant networks formed during epileptogenesis. This review explores the alterations in microglial and astrocytic function and their mechanisms of communication following an epileptogenic insult, examining their contribution to epilepsy development. By comprehensively studying these mechanisms, potential avenues could emerge for refining therapeutic strategies and ameliorating the impact of this complex neurological disease.
Collapse
Affiliation(s)
- Peravina Thergarajan
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Victoria, 3004, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Victoria, 3000, Australia
| | - Nigel C Jones
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Victoria, 3004, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Victoria, 3000, Australia
| | - Idrish Ali
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Victoria, 3004, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Victoria, 3000, Australia
| |
Collapse
|
6
|
Ashtiani SH, Akel S, Berger E, Zelano J. Plasma proteomics in epilepsy: Network-based identification of proteins associated with seizures. Epilepsy Res 2025; 209:107480. [PMID: 39626528 DOI: 10.1016/j.eplepsyres.2024.107480] [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: 04/22/2024] [Revised: 10/31/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE Identification of potential biomarkers of seizures. METHODS In this exploratory study, we quantified plasma protein intensities in 15 patients with recent seizures compared to 15 patients with long-standing seizure freedom. Using TMT-based proteomics we found fifty-one differentially expressed proteins. RESULTS Network analyses including co-expression networks and protein-protein interaction networks, using the STRING database, followed by network centrality and modularity analyses revealed 22 protein modules, with one module showing a significant association with seizures. The protein-protein interaction network centered around this module identified a subnetwork of 125 proteins, grouped into four clusters. Notably, one cluster (mainly enriching inflammatory pathways and Gene Ontology terms) demonstrated the highest enrichment of known epilepsy-related genes. CONCLUSION Overall, our network-based approach identified a protein module linked with seizures. The module contained known markers of epilepsy and inflammation. The results also demonstrate the potential of network analysis in discovering new biomarkers for improved epilepsy management.
Collapse
Affiliation(s)
- Saman Hosseini Ashtiani
- Department of Clinical Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden; Wallenberg Center of Molecular and Translational Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden.
| | - Sarah Akel
- Department of Clinical Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden; Wallenberg Center of Molecular and Translational Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Evelin Berger
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Zelano
- Department of Clinical Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden; Wallenberg Center of Molecular and Translational Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden; Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
7
|
Kwon D, Kim Y, Cho SH. Antidepressant Effects of Ginsenoside Rc on L-Alpha-Aminoadipic Acid-Induced Astrocytic Ablation and Neuroinflammation in Mice. Int J Mol Sci 2024; 25:9673. [PMID: 39273621 PMCID: PMC11396248 DOI: 10.3390/ijms25179673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Depression is a prevalent and debilitating mental disorder that affects millions worldwide. Current treatments, such as antidepressants targeting the serotonergic system, have limitations, including delayed onset of action and high rates of treatment resistance, necessitating novel therapeutic strategies. Ginsenoside Rc (G-Rc) has shown potential anti-inflammatory and neuroprotective effects, but its antidepressant properties remain unexplored. This study investigated the antidepressant effects of G-Rc in an L-alpha-aminoadipic acid (L-AAA)-induced mouse model of depression, which mimics the astrocytic pathology and neuroinflammation observed in major depressive disorder. Mice were administered G-Rc, vehicle, or imipramine orally after L-AAA injection into the prefrontal cortex. G-Rc significantly reduced the immobility time in forced swimming and tail suspension tests compared to vehicle treatment, with more pronounced effects than imipramine. It also attenuated the expression of pro-inflammatory cytokines (TNF-α, IL-6, TGF-β, lipocalin-2) and alleviated astrocytic degeneration, as indicated by increased GFAP and decreased IBA-1 levels. Additionally, G-Rc modulated apoptosis-related proteins, decreasing caspase-3 and increasing Bcl-2 levels compared to the L-AAA-treated group. These findings suggest that G-Rc exerts antidepressant effects by regulating neuroinflammation, astrocyte-microglia crosstalk, and apoptotic pathways in the prefrontal cortex, highlighting its potential as a novel therapeutic agent for depression.
Collapse
Affiliation(s)
- Dohyung Kwon
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yunna Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung-Hun Cho
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
8
|
Laohavisudhi K, Sriwichaiin S, Attachaipanich T, Wittayachamnankul B, Chattipakorn N, Chattipakorn S. Mechanistic insights into Lipocalin-2 in ischemic stroke and hemorrhagic brain injury: Integrating animal and clinical studies. Exp Neurol 2024; 379:114885. [PMID: 38996863 DOI: 10.1016/j.expneurol.2024.114885] [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: 05/06/2024] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Brain injuries, including strokes and traumatic brain injuries (TBI), are a major global health concern, contributing significantly to both mortality and long-term disability. Recent research has identified lipocalin-2 (LCN2), a glycoprotein secreted by various brain cells, as a key factor in influencing brain injury outcomes. Evidence from animal and clinical studies firmly establishes the pivotal role of LCN2 in driving the inflammatory responses triggered by damage to brain tissue. Furthermore, increased LCN2 promotes cellular differentiation, blood-brain barrier breakdown, and decreases cell viability. Interventions with LCN2 inhibitors attenuated brain injury through a reduction in the inflammation process and enhanced cellular viability. Potential mechanisms of LCN2 involve several pathways including the Janus kinase-2 (JAK2)-signal transducers and the transcription-3 (STAT3) signaling, hypoxia-inducible factor 1-alpha (HIF-1α)-LCN2-vascular endothelial growth factor alpha (VEGFα), and the PKR-like ER kinase (PERK) pathways. LCN2 itself interacts with diverse inflammatory cytokines in TBI and intracranial hemorrhage (ICH), resulting in disruption of the blood-brain barrier, increased programmed cell death, and an imbalance in iron homeostasis. Clinical studies have also shown that increased LCN2 level can act as a prognostic biomarker of outcomes following brain injuries. Therefore, this review aims to comprehensively evaluate the role and underlying mechanisms of LCN2 in brain injuries, including stroke and TBI, and explore potential therapeutic interventions targeting LCN2 in these conditions.
Collapse
Affiliation(s)
- Korsin Laohavisudhi
- Department of Emergency Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirawit Sriwichaiin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Research Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Tanawat Attachaipanich
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Borwon Wittayachamnankul
- Department of Emergency Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Research Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
9
|
Qiu R, Cai Y, Su Y, Fan K, Sun Z, Zhang Y. Emerging insights into Lipocalin-2: Unraveling its role in Parkinson's Disease. Biomed Pharmacother 2024; 177:116947. [PMID: 38901198 DOI: 10.1016/j.biopha.2024.116947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 06/22/2024] Open
Abstract
Parkinson's disease (PD) ranks as the second most prevalent neurodegenerative disorder globally, marked by a complex pathogenesis. Lipocalin-2 (LCN2) emerges as a crucial factor during the progression of PD. Belonging to the lipocalin family, LCN2 is integral to several biological functions, including glial cell activation, iron homeostasis regulation, immune response, inflammatory reactions, and oxidative stress mitigation. Substantial research has highlighted marked increases in LCN2 expression within the substantia nigra (SN), cerebrospinal fluid (CSF), and blood of individuals with PD. This review focuses on the pathological roles of LCN2 in neuroinflammation, aging, neuronal damage, and iron dysregulation in PD. It aims to explore the underlying mechanisms of LCN2 in the disease and potential therapeutic targets that could inform future treatment strategies.
Collapse
Affiliation(s)
- Ruqing Qiu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yunjia Cai
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yana Su
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Kangli Fan
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
10
|
Afridi R, Kim JH, Bhusal A, Lee WH, Suk K. Lipocalin-2 as a mediator of neuroimmune communication. J Leukoc Biol 2024; 116:357-368. [PMID: 38149462 DOI: 10.1093/jleuko/qiad157] [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: 10/06/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Lipocalin-2, a neutrophil gelatinase-associated lipocalin, is a 25-kDa secreted protein implicated in a broad range of inflammatory diseases affecting the brain and periphery. It is a pleotropic protein expressed by various immune and nonimmune cells throughout the body. Importantly, the surge in lipocalin-2 levels in disease states has been associated with a myriad of undesirable effects, further exacerbating the ongoing pathological processes. In the brain, glial cells are the principal source of lipocalin-2, which plays a definitive role in determining their functional phenotypes. In different central nervous system pathologies, an increased expression of glial lipocalin-2 has been linked to neurotoxicity. Lipocalin-2 mediates a crosstalk between central and peripheral immune cells under neuroinflammatory conditions. One intriguing aspect is that elevated lipocalin-2 levels in peripheral disorders, such as cancer, metabolic conditions, and liver diseases, potentially incite an inflammatory activation of glial cells while disrupting neuronal functions. This review comprehensively summarizes the influence of lipocalin-2 on the exacerbation of neuroinflammation by regulating various cellular processes. Additionally, this review explores lipocalin-2 as a mediator of neuroimmune crosstalk in various central nervous system pathologies and highlights the role of lipocalin-2 in carrying inflammatory signals along the neuroimmune axis.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41940, Republic of Korea
| | - Jae-Hong Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41940, Republic of Korea
| | - Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41940, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41940, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
| |
Collapse
|
11
|
Gardner RS, Kyle M, Hughes K, Zhao LR. Single-Cell RNA Sequencing Reveals Immunomodulatory Effects of Stem Cell Factor and Granulocyte Colony-Stimulating Factor Treatment in the Brains of Aged APP/PS1 Mice. Biomolecules 2024; 14:827. [PMID: 39062541 PMCID: PMC11275138 DOI: 10.3390/biom14070827] [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: 05/09/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) leads to progressive neurodegeneration and dementia. AD primarily affects older adults with neuropathological changes including amyloid-beta (Aβ) deposition, neuroinflammation, and neurodegeneration. We have previously demonstrated that systemic treatment with combined stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) (SCF+G-CSF) reduces the Aβ load, increases Aβ uptake by activated microglia and macrophages, reduces neuroinflammation, and restores dendrites and synapses in the brains of aged APPswe/PS1dE9 (APP/PS1) mice. However, the mechanisms underlying SCF+G-CSF-enhanced brain repair in aged APP/PS1 mice remain unclear. This study used a transcriptomic approach to identify the potential mechanisms by which SCF+G-CSF treatment modulates microglia and peripheral myeloid cells to mitigate AD pathology in the aged brain. After injections of SCF+G-CSF for 5 consecutive days, single-cell RNA sequencing was performed on CD11b+ cells isolated from the brains of 28-month-old APP/PS1 mice. The vast majority of cell clusters aligned with transcriptional profiles of microglia in various activation states. However, SCF+G-CSF treatment dramatically increased a cell population showing upregulation of marker genes related to peripheral myeloid cells. Flow cytometry data also revealed an SCF+G-CSF-induced increase of cerebral CD45high/CD11b+ active phagocytes. SCF+G-CSF treatment robustly increased the transcription of genes implicated in immune cell activation, including gene sets that regulate inflammatory processes and cell migration. The expression of S100a8 and S100a9 was robustly enhanced following SCF+G-CSF treatment in all CD11b+ cell clusters. Moreover, the topmost genes differentially expressed with SCF+G-CSF treatment were largely upregulated in S100a8/9-positive cells, suggesting a well-conserved transcriptional profile related to SCF+G-CSF treatment in resident and peripherally derived CD11b+ immune cells. This S100a8/9-associated transcriptional profile contained notable genes related to pro-inflammatory and anti-inflammatory responses, neuroprotection, and Aβ plaque inhibition or clearance. Altogether, this study reveals the immunomodulatory effects of SCF+G-CSF treatment in the aged brain with AD pathology, which will guide future studies to further uncover the therapeutic mechanisms.
Collapse
Affiliation(s)
| | | | | | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, 750 E. Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
12
|
Kim JH, Michiko N, Choi IS, Kim Y, Jeong JY, Lee MG, Jang IS, Suk K. Aberrant activation of hippocampal astrocytes causes neuroinflammation and cognitive decline in mice. PLoS Biol 2024; 22:e3002687. [PMID: 38991663 PMCID: PMC11239238 DOI: 10.1371/journal.pbio.3002687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 05/21/2024] [Indexed: 07/13/2024] Open
Abstract
Reactive astrocytes are associated with neuroinflammation and cognitive decline in diverse neuropathologies; however, the underlying mechanisms are unclear. We used optogenetic and chemogenetic tools to identify the crucial roles of the hippocampal CA1 astrocytes in cognitive decline. Our results showed that repeated optogenetic stimulation of the hippocampal CA1 astrocytes induced cognitive impairment in mice and decreased synaptic long-term potentiation (LTP), which was accompanied by the appearance of inflammatory astrocytes. Mechanistic studies conducted using knockout animal models and hippocampal neuronal cultures showed that lipocalin-2 (LCN2), derived from reactive astrocytes, mediated neuroinflammation and induced cognitive impairment by decreasing the LTP through the reduction of neuronal NMDA receptors. Sustained chemogenetic stimulation of hippocampal astrocytes provided similar results. Conversely, these phenomena were attenuated by a metabolic inhibitor of astrocytes. Fiber photometry using GCaMP revealed a high level of hippocampal astrocyte activation in the neuroinflammation model. Our findings suggest that reactive astrocytes in the hippocampus are sufficient and required to induce cognitive decline through LCN2 release and synaptic modulation. This abnormal glial-neuron interaction may contribute to the pathogenesis of cognitive disturbances in neuroinflammation-associated brain conditions.
Collapse
Affiliation(s)
- Jae-Hong Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu, Republic of Korea
| | - Nakamura Michiko
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - In-Sun Choi
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Yujung Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ji-Young Jeong
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Maan-Gee Lee
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Il-Sung Jang
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
13
|
Gardner R, Kyle M, Hughes K, Zhao LR. Single cell RNA sequencing reveals immunomodulatory effects of stem cell factor and granulocyte colony-stimulating factor treatment in the brains of aged APP/PS1 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593359. [PMID: 38766064 PMCID: PMC11100789 DOI: 10.1101/2024.05.09.593359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Alzheimers disease leads to progressive neurodegeneration and dementia. Alzheimers disease primarily affects older adults with neuropathological changes including amyloid beta deposition, neuroinflammation, and neurodegeneration. We have previously demonstrated that systemic treatment with combined stem cell factor, SCF, and granulocyte colony stimulating factor, GCSF, reduces amyloid beta load, increases amyloid beta uptake by activated microglia and macrophages, reduces neuroinflammation, and restores dendrites and synapses in the brains of aged APP-PS1 mice. However, the mechanisms underlying SCF-GCSF-enhanced brain repair in aged APP-PS1 mice remain unclear. This study used a transcriptomic approach to identify potential mechanisms by which SCF-GCSF treatment modulates microglia and peripheral myeloid cells to mitigate Alzheimers disease pathology in the aged brain. After injections of SCF-GCSF for 5 consecutive days, single cell RNA sequencing was performed on CD11b positive cells isolated from the brains of 28-month-old APP-PS1 mice. The vast majority of cell clusters aligned with transcriptional profiles of microglia in various activation states. However, SCF-GCSF treatment dramatically increased a cell population showing upregulation of marker genes related to peripheral myeloid cells. Flow cytometry data also revealed an SCF-GCSF-induced increase of cerebral CD45high-CD11b positive active phagocytes. SCF-GCSF treatment robustly increased the transcription of genes implicated in immune cell activation, including gene sets that regulate inflammatory processes and cell migration. Expression of S100a8 and S100a9 were robustly enhanced following SCF-GCSF treatment in all CD11b positive cell clusters. Moreover, the topmost genes differentially expressed with SCF-GCSF treatment were largely upregulated in S100a8-S100a9 positive cells, suggesting a well-conserved transcriptional profile related to SCF-GCSF treatment in resident and peripherally derived CD11b positive immune cells. This S100a8-S100a9-associated transcriptional profile contained notable genes related to proinflammatory and antiinflammatory responses, neuroprotection, and amyloid beta plaque inhibition or clearance. Altogether, this study reveals immunomodulatory effects of SCF-GCSF treatment in the aged brain with Alzheimers disease pathology, which will guide future studies to further uncover the therapeutic mechanisms.
Collapse
|
14
|
Zhang J, Song Z, Huo Y, Li G, Lu L, Wei C, Zhang S, Gao X, Jiang X, Xu Y. Engeletin alleviates depressive-like behaviours by modulating microglial polarization via the LCN2/CXCL10 signalling pathway. J Cell Mol Med 2024; 28:e18285. [PMID: 38597406 PMCID: PMC11005460 DOI: 10.1111/jcmm.18285] [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: 01/24/2024] [Revised: 03/10/2024] [Accepted: 03/22/2024] [Indexed: 04/11/2024] Open
Abstract
Microglial polarization and associated inflammatory activity are the key mediators of depression pathogenesis. The natural Smilax glabra rhizomilax derivative engeletin has been reported to exhibit robust anti-inflammatory activity, but no studies to date have examined the mechanisms through which it can treat depressive symptoms. We showed that treatment for 21 days with engeletin significantly alleviated depressive-like behaviours in chronic stress social defeat stress (CSDS) model mice. T1-weighted imaging (T1WI), T2-weighted imaging (T2WI) imaging revealed no significant differences between groups, but the bilateral prefrontal cortex of CSDS mice exhibited significant increases in apparent diffusion coefficient and T2 values relative to normal control mice, with a corresponding reduction in fractional anisotropy, while engeletin reversed all of these changes. CSDS resulted in higher levels of IL-1β, IL-6, and TNF-a production, enhanced microglial activation, and greater M1 polarization with a concomitant decrease in M2 polarization in the mPFC, whereas engeletin treatment effectively abrogated these CSDS-related pathological changes. Engeletin was further found to suppress the LCN2/C-X-C motif chemokine ligand 10 (CXCL10) signalling axis such that adeno-associated virus-induced LCN2 overexpression ablated the antidepressant effects of engeletin and reversed its beneficial effects on the M1/M2 polarization of microglia. In conclusion, engeletin can alleviate CSDS-induced depressive-like behaviours by regulating the LCN2/CXCL10 pathway and thereby altering the polarization of microglia. These data suggest that the antidepressant effects of engeletin are correlated with the polarization of microglia, highlighting a potential avenue for future design of antidepressant strategies that specifically target the microglia.
Collapse
Affiliation(s)
- Jie Zhang
- Department of RadiologyBinzhou Medical University HospitalBinzhouShandongP. R. China
| | - Zheng Song
- Department of PharmacyBinzhou Medical University HospitalBinzhouShandongP. R. China
| | - Yanchao Huo
- Department of PharmacyBinzhou Medical University HospitalBinzhouShandongP. R. China
| | - Guangqiang Li
- Department of PharmacyBinzhou Medical University HospitalBinzhouShandongP. R. China
| | - Liming Lu
- Department of PharmacyBinzhou Medical University HospitalBinzhouShandongP. R. China
| | - Chuanmei Wei
- Department of PharmacyBinzhou Medical University HospitalBinzhouShandongP. R. China
| | - Shuping Zhang
- College of Basic MedicineBinzhou Medical UniversityYantaiShandongP.R. China
| | - Xinfu Gao
- Department of PharmacyBinzhou Medical University HospitalBinzhouShandongP. R. China
| | - Xingyue Jiang
- Department of RadiologyBinzhou Medical University HospitalBinzhouShandongP. R. China
| | - Yangyang Xu
- Department of PharmacyBinzhou Medical University HospitalBinzhouShandongP. R. China
| |
Collapse
|
15
|
Abcouwer SF, Miglioranza Scavuzzi B, Kish PE, Kong D, Shanmugam S, Le XA, Yao J, Hager H, Zacks DN. The mouse retinal pigment epithelium mounts an innate immune defense response following retinal detachment. J Neuroinflammation 2024; 21:74. [PMID: 38528525 PMCID: PMC10964713 DOI: 10.1186/s12974-024-03062-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/11/2024] [Indexed: 03/27/2024] Open
Abstract
The retinal pigment epithelium (RPE) maintains photoreceptor viability and function, completes the visual cycle, and forms the outer blood-retinal barrier (oBRB). Loss of RPE function gives rise to several monogenic retinal dystrophies and contributes to age-related macular degeneration. Retinal detachment (RD) causes separation of the neurosensory retina from the underlying RPE, disrupting the functional and metabolic relationships between these layers. Although the retinal response to RD is highly studied, little is known about how the RPE responds to loss of this interaction. RNA sequencing (RNA-Seq) was used to compare normal and detached RPE in the C57BL6/J mouse. The naïve mouse RPE transcriptome was compared to previously published RPE signature gene lists and from the union of these 14 genes (Bmp4, Crim1, Degs1, Gja1, Itgav, Mfap3l, Pdpn, Ptgds, Rbp1, Rnf13, Rpe65, Slc4a2, Sulf1 and Ttr) representing a core signature gene set applicable across rodent and human RPE was derived. Gene ontology enrichment analysis (GOEA) of the mouse RPE transcriptome identified expected RPE features and functions, such as pigmentation, phagocytosis, lysosomal and proteasomal degradation of proteins, and barrier function. Differentially expressed genes (DEG) at 1 and 7 days post retinal detachment (dprd) were defined as mRNA with a significant (padj≤0.05) fold change (FC) of 0.67 ≥ FC ≥ 1.5 in detached versus naïve RPE. The RPE transcriptome exhibited dramatic changes at 1 dprd, with 2297 DEG identified. The KEGG pathways and biological process GO groups related to innate immune responses were significantly enriched. Lipocalin 2 (Lcn2) and several chemokines were upregulated, while numerous genes related to RPE functions, such as pigment synthesis, visual cycle, phagocytosis, and tight junctions were downregulated at 1 dprd. The response was largely transient, with only 18 significant DEG identified at 7 dprd, including upregulation of complement gene C4b. Validation studies confirmed RNA-Seq results. Thus, the RPE quickly downregulates cell-specific functions and mounts an innate immune defense response following RD. Our data demonstrate that the RPE contributes to the inflammatory response to RD and may play a role in attraction of immune cells to the subretinal space.
Collapse
Affiliation(s)
- Steven F Abcouwer
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA.
| | - Bruna Miglioranza Scavuzzi
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - Phillip E Kish
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - Dejuan Kong
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - Sumathi Shanmugam
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - Xuan An Le
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - Jingyu Yao
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - Heather Hager
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - David N Zacks
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| |
Collapse
|
16
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
17
|
Salem NA, Manzano L, Keist MW, Ponomareva O, Roberts AJ, Roberto M, Mayfield RD. Cell-type brain-region specific changes in prefrontal cortex of a mouse model of alcohol dependence. Neurobiol Dis 2024; 190:106361. [PMID: 37992784 PMCID: PMC10874299 DOI: 10.1016/j.nbd.2023.106361] [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: 06/29/2023] [Revised: 10/31/2023] [Accepted: 11/18/2023] [Indexed: 11/24/2023] Open
Abstract
The prefrontal cortex is a crucial regulator of alcohol drinking, and dependence, and other behavioral phenotypes associated with AUD. Comprehensive identification of cell-type specific transcriptomic changes in alcohol dependence will improve our understanding of mechanisms underlying the excessive alcohol use associated with alcohol dependence and will refine targets for therapeutic development. We performed single nucleus RNA sequencing (snRNA-seq) and Visium spatial gene expression profiling on the medial prefrontal cortex (mPFC) obtained from C57BL/6 J mice exposed to the two-bottle choice-chronic intermittent ethanol (CIE) vapor exposure (2BC-CIE, defined as dependent group) paradigm which models phenotypes of alcohol dependence including escalation of alcohol drinking. Gene co-expression network analysis and differential expression analysis identified highly dysregulated co-expression networks in multiple cell types. Dysregulated modules and their hub genes suggest novel understudied targets for studying molecular mechanisms contributing to the alcohol dependence state. A subtype of inhibitory neurons was the most alcohol-sensitive cell type and contained a downregulated gene co-expression module; the hub gene for this module is Cpa6, a gene previously identified by GWAS to be associated with excessive alcohol consumption. We identified an astrocytic Gpc5 module significantly upregulated in the alcohol-dependent group. To our knowledge, there are no studies linking Cpa6 and Gpc5 to the alcohol-dependent phenotype. We also identified neuroinflammation related gene expression changes in multiple cell types, specifically enriched in microglia, further implicating neuroinflammation in the escalation of alcohol drinking. Here, we present a comprehensive atlas of cell-type specific alcohol dependence mediated gene expression changes in the mPFC and identify novel cell type-specific targets implicated in alcohol dependence.
Collapse
Affiliation(s)
- Nihal A Salem
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Lawrence Manzano
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Michael W Keist
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Olga Ponomareva
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
18
|
Sciarretta F, Ceci V, Tiberi M, Zaccaria F, Li H, Zhou ZY, Sun Q, Konja D, Matteocci A, Bhusal A, Verri M, Fresegna D, Balletta S, Ninni A, Di Biagio C, Rosina M, Suk K, Centonze D, Wang Y, Chiurchiù V, Aquilano K, Lettieri-Barbato D. Lipocalin-2 promotes adipose-macrophage interactions to shape peripheral and central inflammatory responses in experimental autoimmune encephalomyelitis. Mol Metab 2023; 76:101783. [PMID: 37517520 PMCID: PMC10448472 DOI: 10.1016/j.molmet.2023.101783] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023] Open
Abstract
OBJECTIVE Accumulating evidence suggests that dysfunctional adipose tissue (AT) plays a major role in the risk of developing multiple sclerosis (MS), the most common immune-mediated and demyelinating disease of the central nervous system. However, the contribution of adipose tissue to the etiology and progression of MS is still obscure. This study aimed at deciphering the responses of AT in experimental autoimmune encephalomyelitis (EAE), the best characterized animal model of MS. RESULTS AND METHODS We observed a significant AT loss in EAE mice at the onset of disease, with a significant infiltration of M1-like macrophages and fibrosis in the AT, resembling a cachectic phenotype. Through an integrative and multilayered approach, we identified lipocalin2 (LCN2) as the key molecule released by dysfunctional adipocytes through redox-dependent mechanism. Adipose-derived LCN2 shapes the pro-inflammatory macrophage phenotype, and the genetic deficiency of LCN2 specifically in AT reduced weight loss as well as inflammatory macrophage infiltration in spinal cord in EAE mice. Mature adipocytes downregulating LCN2 reduced lipolytic response to inflammatory stimuli (e.g. TNFα) through an ATGL-mediated mechanism. CONCLUSIONS Overall data highlighted a role LCN2 in exacerbating inflammatory phenotype in EAE model, suggesting a pathogenic role of dysfunctional AT in MS.
Collapse
Affiliation(s)
| | - Veronica Ceci
- PhD Program in Evolutionary Biology and Ecology, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marta Tiberi
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Fabio Zaccaria
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Haoyun Li
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Zhong-Yan Zhou
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiyang Sun
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; PhD program in Immunology, Molecular Medicine and Applied biotechnologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Martina Verri
- Pathology Unit, University Hospital Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00163 Rome, Italy
| | - Sara Balletta
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Andrea Ninni
- PhD Program in Evolutionary Biology and Ecology, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Claudia Di Biagio
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marco Rosina
- Neurology Unit, Fondazione PTV Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Daniele Lettieri-Barbato
- IRCCS, Fondazione Santa Lucia, 00179 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy.
| |
Collapse
|
19
|
Zelenka L, Jarek M, Pägelow D, Geffers R, van Vorst K, Fulde M. Crosstalk of Highly Purified Microglia and Astrocytes in the Frame of Toll-like Receptor (TLR)2/1 Activation. Neuroscience 2023; 526:256-266. [PMID: 37391121 DOI: 10.1016/j.neuroscience.2023.05.001] [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: 07/21/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 07/02/2023]
Abstract
The major immune cells of the central nervous systems (CNS) are microglia and astrocytes, subsets of the glial cell population. The crosstalk between glia via soluble signaling molecules plays an indispensable role for neuropathologies, brain development as well as homeostasis. However, the investigation of the microglia-astrocyte crosstalk has been hampered due to the lack of suitable glial isolation methods. In this study, we investigated for the first time the crosstalk between highly purified Toll-like receptor (TLR)2-knock out (TLR2-KO) and wild-type (WT) microglia and astrocytes. We examined the crosstalk of TLR2-KO microglia and astrocytes in the presence of WT supernatants of the respective other glial cell type. Interestingly, we observed a significant TNF release by TLR2-KO astrocytes, which were activated with Pam3CSK4-stimulated WT microglial supernatants, strongly indicating a crosstalk between microglia and astrocytes after TLR2/1 activation. Furthermore, transcriptome analysis using RNA-seq revealed a wide range of significant up- and down-regulated genes such as Cd300, Tnfrsf9 or Lcn2, which might be involved in the molecular conversation between microglia and astrocytes. Finally, co-culturing microglia and astrocytes confirmed the prior results by demonstrating a significant TNF release by WT microglia co-cultured with TLR2-KO astrocytes. Our findings suggest a molecular TLR2/1-dependent conversation between highly pure activated microglia and astrocytes via signaling molecules. Furthermore, we demonstrate the first crosstalk experiments using ∼100% pure microglia and astrocyte mono-/co-cultures derived from mice with different genotypes highlighting the urgent need of efficient glial isolation protocols, which particularly holds true for astrocytes.
Collapse
Affiliation(s)
- Laura Zelenka
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Michael Jarek
- Helmholtz Centre for Infection Research, Research Group Genome Analytics (GMAK), Braunschweig, Germany
| | - Dennis Pägelow
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Robert Geffers
- Helmholtz Centre for Infection Research, Research Group Genome Analytics (GMAK), Braunschweig, Germany
| | - Kira van Vorst
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Marcus Fulde
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
20
|
Chen Y, Zheng D, Wang H, Zhang S, Zhou Y, Ke X, Chen G. Lipocalin 2 in the Paraventricular Thalamic Nucleus Contributes to DSS-Induced Depressive-Like Behaviors. Neurosci Bull 2023; 39:1263-1277. [PMID: 36920644 PMCID: PMC10387009 DOI: 10.1007/s12264-023-01047-4] [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: 09/21/2022] [Accepted: 01/23/2023] [Indexed: 03/16/2023] Open
Abstract
The incidence rate of anxiety and depression is significantly higher in patients with inflammatory bowel diseases (IBD) than in the general population. The mechanisms underlying dextran sulfate sodium (DSS)-induced depressive-like behaviors are still unclear. We clarified that IBD mice induced by repeated administration of DSS presented depressive-like behaviors. The paraventricular thalamic nucleus (PVT) was regarded as the activated brain region by the number of c-fos-labeled neurons. RNA-sequencing analysis showed that lipocalin 2 (Lcn2) was upregulated in the PVT of mice with DSS-induced depressive behaviors. Upregulating Lcn2 from neuronal activity induced dendritic spine loss and the secreted protein induced chemokine expression and subsequently contributed to microglial activation leading to blood-brain barrier permeability. Moreover, Lcn2 silencing in the PVT alleviated the DSS-induced depressive-like behaviors. The present study demonstrated that elevated Lcn2 in the PVT is a critical factor for DSS-induced depressive behaviors.
Collapse
Affiliation(s)
- Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Du Zheng
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Hongwei Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Shuxia Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Youfa Zhou
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Xinlong Ke
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
21
|
Zheng XQ, Lin JL, Huang J, Wu T, Song CL. Targeting aging with the healthy skeletal system: The endocrine role of bone. Rev Endocr Metab Disord 2023; 24:695-711. [PMID: 37402956 DOI: 10.1007/s11154-023-09812-6] [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] [Accepted: 05/29/2023] [Indexed: 07/06/2023]
Abstract
Aging is an inevitable biological process, and longevity may be related to bone health. Maintaining strong bone health can extend one's lifespan, but the exact mechanism is unclear. Bone and extraosseous organs, including the heart and brain, have complex and precise communication mechanisms. In addition to its load bearing capacity, the skeletal system secretes cytokines, which play a role in bone regulation of extraosseous organs. FGF23, OCN, and LCN2 are three representative bone-derived cytokines involved in energy metabolism, endocrine homeostasis and systemic chronic inflammation levels. Today, advanced research methods provide new understandings of bone as a crucial endocrine organ. For example, gene editing technology enables bone-specific conditional gene knockout models, which allows the study of bone-derived cytokines to be more precise. We systematically evaluated the various effects of bone-derived cytokines on extraosseous organs and their possible antiaging mechanism. Targeting aging with the current knowledge of the healthy skeletal system is a potential therapeutic strategy. Therefore, we present a comprehensive review that summarizes the current knowledge and provides insights for futures studies.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jia-Liang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jie Huang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Tong Wu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China.
| |
Collapse
|
22
|
Xiao M, Li X, Wang L, Lin B, Zhai M, Hull L, Zizzo A, Cui W, Kiang JG. Skin Wound following Irradiation Aggravates Radiation-Induced Brain Injury in a Mouse Model. Int J Mol Sci 2023; 24:10701. [PMID: 37445879 DOI: 10.3390/ijms241310701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Radiation injury- and radiation combined with skin injury-induced inflammatory responses in the mouse brain were evaluated in this study. Female B6D2F1/J mice were subjected to a sham, a skin wound (SW), 9.5 Gy 60Co total-body gamma irradiation (RI), or 9.5 Gy RI combined with a skin puncture wound (RCI). Survival, body weight, and wound healing were tracked for 30 days, and mouse brain samples were collected on day 30 after SW, RI, RCI, and the sham control. Our results showed that RCI caused more severe animal death and body weight loss compared with RI, and skin wound healing was significantly delayed by RCI compared to SW. RCI and RI increased the chemokines Eotaxin, IP-10, MIG, 6Ckine/Exodus2, MCP-5, and TIMP-1 in the brain compared to SW and the sham control mice, and the Western blot results showed that IP-10 and p21 were significantly upregulated in brain cells post-RI or -RCI. RI and RCI activated both astrocytes and endothelial cells in the mouse brain, subsequently inducing blood-brain barrier (BBB) leakage, as shown by the increased ICAM1 and GFAP proteins in the brain and GFAP in the serum. The Doublecortin (DCX) protein, the "gold standard" for measuring neurogenesis, was significantly downregulated by RI and RCI compared with the sham group. Furthermore, RI and RCI decreased the expression of the neural stem cell marker E-cadherin, the intermediate progenitor marker MASH1, the immature neuron cell marker NeuroD1, and the mature neuron cell marker NeuN, indicating neural cell damage in all development stages after RI and RCI. Immunohistochemistry (IHC) staining further confirmed the significant loss of neural cells in RCI. Our data demonstrated that RI and RCI induced brain injury through inflammatory pathways, and RCI exacerbated neural cell damage more than RI.
Collapse
Affiliation(s)
- Mang Xiao
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Xianghong Li
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Li Wang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Bin Lin
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Min Zhai
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Lisa Hull
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Alex Zizzo
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Wanchang Cui
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Juliann G Kiang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
23
|
Zhao RY, Wei PJ, Sun X, Zhang DH, He QY, Liu J, Chang JL, Yang Y, Guo ZN. Role of lipocalin 2 in stroke. Neurobiol Dis 2023; 179:106044. [PMID: 36804285 DOI: 10.1016/j.nbd.2023.106044] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/22/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023] Open
Abstract
Stroke is the second leading cause of death worldwide; however, the treatment choices available to neurologists are limited in clinical practice. Lipocalin 2 (LCN2) is a secreted protein, belonging to the lipocalin superfamily, with multiple biological functions in mediating innate immune response, inflammatory response, iron-homeostasis, cell migration and differentiation, energy metabolism, and other processes in the body. LCN2 is expressed at low levels in the brain under normal physiological conditions, but its expression is significantly up-regulated in multiple acute stimulations and chronic pathologies. An up-regulation of LCN2 has been found in the blood/cerebrospinal fluid of patients with ischemic/hemorrhagic stroke, and could serve as a potential biomarker for the prediction of the severity of acute stroke. LCN2 activates reactive astrocytes and microglia, promotes neutrophil infiltration, amplifies post-stroke inflammation, promotes blood-brain barrier disruption, white matter injury, and neuronal death. Moreover, LCN2 is involved in brain injury induced by thrombin and erythrocyte lysates, as well as microvascular thrombosis after hemorrhage. In this paper, we review the role of LCN2 in the pathological processes of ischemic stroke; intracerebral hemorrhage; subarachnoid hemorrhage; and stroke-related brain diseases, such as vascular dementia and post-stroke depression, and their underlying mechanisms. We hope that this review will help elucidate the value of LCN2 as a therapeutic target in stroke.
Collapse
Affiliation(s)
- Ruo-Yu Zhao
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Peng-Ju Wei
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Sun
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Dian-Hui Zhang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Jie Liu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Jun-Lei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China; Neuroscience Research Center, the First Hospital of Jilin University, Chang Chun, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China; Neuroscience Research Center, the First Hospital of Jilin University, Chang Chun, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China.
| |
Collapse
|
24
|
Kim JH, Kang RJ, Hyeon SJ, Ryu H, Joo H, Bu Y, Kim JH, Suk K. Lipocalin-2 Is a Key Regulator of Neuroinflammation in Secondary Traumatic and Ischemic Brain Injury. Neurotherapeutics 2023; 20:803-821. [PMID: 36508119 PMCID: PMC10275845 DOI: 10.1007/s13311-022-01333-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Reactive glial cells are hallmarks of brain injury. However, whether these cells contribute to secondary inflammatory pathology and neurological deficits remains poorly understood. Lipocalin-2 (LCN2) has inflammatory and neurotoxic effects in various disease models; however, its pathogenic role in traumatic brain injury remains unknown. The aim of the present study was to investigate the expression of LCN2 and its role in neuroinflammation following brain injury. LCN2 expression was high in the mouse brain after controlled cortical impact (CCI) and photothrombotic stroke (PTS) injury. Brain levels of LCN2 mRNA and protein were also significantly higher in patients with chronic traumatic encephalopathy (CTE) than in normal subjects. RT-PCR and immunofluorescence analyses revealed that astrocytes were the major cellular source of LCN2 in the injured brain. Lcn2 deficiency or intracisternal injection of an LCN2 neutralizing antibody reduced CCI- and PTS-induced brain lesions, behavioral deficits, and neuroinflammation. Mechanistically, in cultured glial cells, recombinant LCN2 protein enhanced scratch injury-induced proinflammatory cytokine gene expression and inhibited Gdnf gene expression, whereas Lcn2 deficiency exerted opposite effects. Together, our results from CTE patients, rodent brain injury models, and cultured glial cells suggest that LCN2 mediates secondary damage response to traumatic and ischemic brain injury by promoting neuroinflammation and suppressing the expression of neurotropic factors.
Collapse
Affiliation(s)
- Jae-Hong Kim
- Brain Korea 21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ri Jin Kang
- Brain Korea 21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Jae Hyeon
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hoon Ryu
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Veterans Affairs Boston Healthcare System, Boston, MA USA
- Boston University Alzheimer’s Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA USA
| | - Hyejin Joo
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Present Address: Pharmacological Research Division, Toxicological Evaluation and Research Department, Ministry of Food and Drug Safety, National Institute of Food and Drug Safety Evaluation, Chungju, Republic of Korea
| | - Youngmin Bu
- Department of Herbal Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jong-Heon Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoungho Suk
- Brain Korea 21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
25
|
Adler O, Zait Y, Cohen N, Blazquez R, Doron H, Monteran L, Scharff Y, Shami T, Mundhe D, Glehr G, Kanner AA, Horn S, Yahalom V, Haferkamp S, Hutchinson JA, Bleckmann A, Nahary L, Benhar I, Yust Katz S, Pukrop T, Erez N. Reciprocal interactions between innate immune cells and astrocytes facilitate neuroinflammation and brain metastasis via lipocalin-2. NATURE CANCER 2023; 4:401-418. [PMID: 36797502 DOI: 10.1038/s43018-023-00519-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/17/2023] [Indexed: 04/19/2023]
Abstract
Brain metastasis still encompass very grim prognosis and therefore understanding the underlying mechanisms is an urgent need toward developing better therapeutic strategies. We uncover the intricate interactions between recruited innate immune cells and resident astrocytes in the brain metastatic niche that facilitate metastasis of melanoma and breast cancer. We show that granulocyte-derived lipocalin-2 (LCN2) induces inflammatory activation of astrocytes, leading to myeloid cell recruitment to the brain. LCN2 is central to inducing neuroinflammation as its genetic targeting or bone-marrow transplantation from LCN2-/- mice was sufficient to attenuate neuroinflammation and inhibit brain metastasis. Moreover, high LCN2 levels in patient blood and brain metastases in multiple cancer types were strongly associated with disease progression and poor survival. Our findings uncover a previously unknown mechanism, establishing a central role for the reciprocal interactions between granulocytes and astrocytes in promoting brain metastasis and implicate LCN2 as a prognostic marker and potential therapeutic target.
Collapse
Affiliation(s)
- Omer Adler
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Zait
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noam Cohen
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Raquel Blazquez
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Hila Doron
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lea Monteran
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yeela Scharff
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Shami
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dhanashree Mundhe
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gunther Glehr
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Andrew A Kanner
- Department of Neurosurgery, Rabin Medical Center and Sackler Faculty of Medicine Tel Aviv University, Tel Aviv, Israel
| | - Suzana Horn
- Department of Pathology, Rabin Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vered Yahalom
- Blood Services & Apheresis Institute, Rabin Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Annalen Bleckmann
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
- Medical Clinic A, Haematology, Haemostasiology, Oncology and Pulmonology, University Hospital Münster, Münster, Germany
- West German Cancer Center, University Hospital Münster, Münster, Germany
| | - Limor Nahary
- The Shmunis School of Biomedicine and Cancer Research, the George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Itai Benhar
- The Shmunis School of Biomedicine and Cancer Research, the George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shlomit Yust Katz
- Neuro-Oncology Unit, Davidoff Cancer Center at Rabin Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Regensburg, Germany
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
26
|
Role of Zerumbone, a Phytochemical Sesquiterpenoid from Zingiber zerumbet Smith, in Maintaining Macrophage Polarization and Redox Homeostasis. Nutrients 2022; 14:nu14245402. [PMID: 36558562 PMCID: PMC9783216 DOI: 10.3390/nu14245402] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Macrophages and microglia are highly versatile cells that can be polarized into M1 and M2 phenotypes in response to diverse environmental stimuli, thus exhibiting different biological functions. In the central nervous system, activated resident macrophages and microglial cells trigger the production of proinflammatory mediators that contribute to neurodegenerative diseases and psychiatric disorders. Therefore, modulating the activation of macrophages and microglia by optimizing the inflammatory environment is beneficial for disease management. Several naturally occurring compounds have been reported to have anti-inflammatory and neuroprotective properties. Zerumbone is a phytochemical sesquiterpenoid and also a cyclic ketone isolated from Zingiber zerumbet Smith. In this study, we found that zerumbone effectively reduced the expression of lipocalin-2 in macrophages and microglial cell lines. Lipocalin-2, also known as neutrophil gelatinase-associated lipocalin (NGAL), has been characterized as an adipokine/cytokine implicated in inflammation. Moreover, supplement with zerumbone inhibited reactive oxygen species production. Phagocytic activity was decreased following the zerumbone supplement. In addition, the zerumbone supplement remarkably reduced the production of M1-polarization-associated chemokines CXC10 and CCL-2, as well as M1-polarization-associated cytokines interleukin (IL)-6, IL-1β, and tumor necrosis factor-α. Furthermore, the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 and the production of NO were attenuated in macrophages and microglial cells supplemented with zerumbone. Notably, we discovered that zerumbone effectively promoted the production of the endogenous antioxidants heme oxygenase-1, glutamate-cysteine ligase modifier subunit, glutamate-cysteine ligase catalytic subunit, and NAD(P)H quinone oxidoreductase-1 and remarkably enhanced IL-10, a marker of M2 macrophage polarization. Endogenous antioxidant production and M2 macrophage polarization were increased through activation of the AMPK/Akt and Akt/GSK3 signaling pathways. In summary, this study demonstrated the protective role of zerumbone in maintaining M1 and M2 polarization homeostasis by decreasing inflammatory responses and enhancing the production of endogenous antioxidants in both macrophages and microglia cells. This study suggests that zerumbone can be used as a potential therapeutic drug for the supplement of neuroinflammatory diseases.
Collapse
|
27
|
Zhang J, Wang Z, Zhang H, Li S, Li J, Liu H, Cheng Q. The role of lipocalin 2 in brain injury and recovery after ischemic and hemorrhagic stroke. Front Mol Neurosci 2022; 15:930526. [PMID: 36187347 PMCID: PMC9520288 DOI: 10.3389/fnmol.2022.930526] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/02/2022] [Indexed: 12/03/2022] Open
Abstract
Ischemic and hemorrhagic stroke (including intracerebral hemorrhage, intraventricular hemorrhage, and subarachnoid hemorrhage) is the dominating cause of disability and death worldwide. Neuroinflammation, blood-brain barrier (BBB) disruption, neuronal death are the main pathological progress, which eventually causes brain injury. Increasing evidence indicated that lipocalin 2 (LCN2), a 25k-Da acute phase protein from the lipocalin superfamily, significantly increased immediately after the stroke and played a vital role in these events. Meanwhile, there exists a close relationship between LCN2 levels and the worse clinical outcome of patients with stroke. Further research revealed that LCN2 elimination is associated with reduced immune infiltrates, infarct volume, brain edema, BBB leakage, neuronal death, and neurological deficits. However, some studies revealed that LCN2 might also act as a beneficial factor in ischemic stroke. Nevertheless, the specific mechanism of LCN2 and its primary receptors (24p3R and megalin) involving in brain injury remains unclear. Therefore, it is necessary to investigate the mechanism of LCN2 induced brain damage after stroke. This review focuses on the role of LCN2 and its receptors in brain injury and aiming to find out possible therapeutic targets to reduce brain damage following stroke.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Shuwang Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Li
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hongwei Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Clinical Diagnosis and Therapy Center for Glioma of Xiangya Hospital, Central South University, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Gasterich N, Bohn A, Sesterhenn A, Nebelo F, Fein L, Kaddatz H, Nyamoya S, Kant S, Kipp M, Weiskirchen R, Zendedel A, Beyer C, Clarner T. Lipocalin 2 attenuates oligodendrocyte loss and immune cell infiltration in mouse models for multiple sclerosis. Glia 2022; 70:2188-2206. [PMID: 35856297 DOI: 10.1002/glia.24245] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 11/06/2022]
Abstract
Multiple sclerosis (MS) is a central nervous system disease characterized by both degenerative and inflammatory processes. Various mediators are involved in the interplay of degeneration and innate immunity on one hand and peripheral adaptive immunity on the other hand. The secreted protein lipocalin 2 (LCN2) is an inflammatory modulator in a variety of pathologies. Although elevated intrathecal levels of LCN2 have been reported in MS patients, it's functional role is widely unknown. Here, we identified a subpopulation of astrocytes as a source of LCN2 in MS lesions and respective animal models. We investigated the functional role of LCN2 for both autoimmune and degenerative aspects in three MS mouse models including both wild type (WT) and Lcn2-/- mouse strains. While the experimental autoimmune encephalomyelitis (EAE) model reflects primary autoimmunity, the cuprizone model reflects selective oligodendrocyte loss and demyelination. In addition, we included a combinatory Cup/EAE model in which primary cytodegeneration is followed by inflammatory lesions within the forebrain. While in the EAE model, the disease outcome was comparable in between the two mouse strains, cuprizone intoxicated Lcn2-/- animals showed an increased loss of oligodendrocytes. In the Cup/EAE model, Lcn2-/- animals showed increased inflammation when compared to WT mice. Together, our results highlight LCN2 as a potentially protective molecule in MS lesion formation, which might be able to limit loss of oligodendrocytes immune-cell invasion. Despite these findings, it is not yet clear which glial cell phenotype (and to which extent) contributes to the observed neuroprotective effects, that is, microglia and/or astroglia or even endothelial cells in the brain.
Collapse
Affiliation(s)
- Natalie Gasterich
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| | - Amelie Bohn
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| | - Anika Sesterhenn
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| | - Frederik Nebelo
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| | - Lena Fein
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| | - Hannes Kaddatz
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| | - Stella Nyamoya
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| | - Sebastian Kant
- RWTH University Hospital Aachen, Institute of Molecular and Cellular Anatomy, Aachen, Germany
| | - Markus Kipp
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| | - Ralf Weiskirchen
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Aachen, Germany
| | - Adib Zendedel
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| | - Cordian Beyer
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| | - Tim Clarner
- RWTH University Hospital Aachen, Institute of Neuroanatomy, Aachen, Germany
| |
Collapse
|
29
|
Increased Plasma Lipocalin-2 Levels in Patients with Myelin Oligodendrocyte Glycoprotein-IgG–Positive Optic Neuritis. J Clin Med 2022; 11:jcm11092635. [PMID: 35566760 PMCID: PMC9105342 DOI: 10.3390/jcm11092635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/08/2022] [Accepted: 05/05/2022] [Indexed: 12/04/2022] Open
Abstract
This study aimed to evaluate the correlation between plasma lipocalin-2 (LCN2) levels and myelin oligodendrocyte glycoprotein (MOG)-immunoglobulin G (IgG) seropositivity in patients with optic neuritis. Peripheral blood samples were collected from 19 patients with optic neuritis and 20 healthy controls. Plasma LCN2 and MOG-IgG levels were measured using enzyme-linked immunosorbent assay and a cell-based assay, respectively. The correlation between plasma LCN2 levels and MOG-IgG titers in patients with optic neuritis was analyzed. Receiver operating characteristic (ROC) curves were constructed to assess and compare the ability of plasma LCN2 and MOG-IgG levels for predicting optic neuritis recurrence. Patients with MOG-IgG–positive optic neuritis had significantly higher mean plasma LCN2 levels than controls and patients with MOG-IgG–negative optic neuritis (p = 0.037). Plasma LCN2 and MOG-IgG levels were significantly correlated in patients with optic neuritis (r = 0.553, p = 0.0141). There were no significant differences in the areas under the ROC curve (AUC) of plasma LCN2 (0.693, 95% confidence interval [CI] 0.443–0.880, p = 0.133) and MOG-IgG (0.641, 95% CI, 0.400–0.840, p = 0.298) levels (95% CI, −0.266–0.448, p = 0.618). Plasma LCN2 levels may aid differentiation of MOG-IgG–positive optic neuritis from MOG-IgG–negative optic neuritis.
Collapse
|
30
|
Luo C, Zhou S, Yin S, Jian L, Luo P, Dong J, Liu E. Lipocalin-2 and Cerebral Stroke. Front Mol Neurosci 2022; 15:850849. [PMID: 35493318 PMCID: PMC9039332 DOI: 10.3389/fnmol.2022.850849] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/23/2022] [Indexed: 11/26/2022] Open
Abstract
Stroke is a common and devastating disease with an escalating prevalence worldwide. The known secondary injuries after stroke include cell death, neuroinflammation, blood-brain barrier disruption, oxidative stress, iron dysregulation, and neurovascular unit dysfunction. Lipocalin-2 (LCN-2) is a neutrophil gelatinase-associated protein that influences diverse cellular processes during a stroke. The role of LCN-2 has been widely recognized in the peripheral system; however, recent findings have revealed that there are links between LCN-2 and secondary injury and diseases in the central nervous system. Novel roles of LCN-2 in neurons, microglia, astrocytes, and endothelial cells have also been demonstrated. Here, we review the evidence on the regulatory roles of LCN-2 in secondary injuries following a stroke from various perspectives and the pathological mechanisms involved in the modulation of stroke. Overall, our review suggests that LCN-2 is a promising target to promote a better understanding of the neuropathology of stroke.
Collapse
Affiliation(s)
- Chao Luo
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Shuai Zhou
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Neurosurgery, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Shi Yin
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Lipeng Jian
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Pengren Luo
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Jigeng Dong
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Erheng Liu
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
31
|
Astroglial and oligodendroglial markers in the cuprizone animal model for de- and remyelination. Histochem Cell Biol 2022; 158:15-38. [PMID: 35380252 PMCID: PMC9246805 DOI: 10.1007/s00418-022-02096-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2022] [Indexed: 01/08/2023]
Abstract
Myelin loss with consecutive axon degeneration and impaired remyelination are the underlying causes of progressive disease in patients with multiple sclerosis. Astrocytes are suggested to play a major role in these processes. The unmasking of distinct astrocyte identities in health and disease would help to understand the pathophysiological mechanisms in which astrocytes are involved. However, the number of specific astrocyte markers is limited. Therefore, we performed immunohistochemical studies and analyzed various markers including GFAP, vimentin, S100B, ALDH1L1, and LCN2 during de- and remyelination using the toxic murine cuprizone animal model. Applying this animal model, we were able to confirm overlapping expression of vimentin and GFAP and highlighted the potential of ALDH1L1 as a pan-astrocytic marker, in agreement with previous data. Only a small population of GFAP-positive astrocytes in the corpus callosum highly up-regulated LCN2 at the peak of demyelination and S100B expression was found in a subset of oligodendroglia as well, thus S100B turned out to have a limited use as a particular astroglial marker. Additionally, numerous GFAP-positive astrocytes in the lateral corpus callosum did not express S100B, further strengthening findings of heterogeneity in the astrocytic population. In conclusion, our results acknowledged that GFAP, vimentin, LCN2, and ALDH1L1 serve as reliable marker to identify activated astrocytes during cuprizone-induced de- and remyelination. Moreover, there were clear regional and temporal differences in protein and mRNA expression levels and patterns of the studied markers, generally between gray and white matter structures.
Collapse
|
32
|
Circulating Lipocalin-2 level is positively associated with cognitive impairment in patients with metabolic syndrome. Sci Rep 2022; 12:4635. [PMID: 35302058 PMCID: PMC8931051 DOI: 10.1038/s41598-022-08286-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/04/2022] [Indexed: 02/07/2023] Open
Abstract
The association between Lipocalin-2 (LCN2) and cognition in patients with metabolic syndrome (MetS) has not been thoroughly investigated. We aimed to evaluate whether serum LCN2 levels are associated with the alteration of cognitive function in patients with MetS. The total of 191 non-demented participants with MetS were enrolled onto the study in 2015, and a cohort study was conducted in a subpopulation in 2020. After adjustment for sex, age, waist circumference, creatinine levels, and HbA1C, an association between the higher serum LCN2 levels and the lower Montreal cognitive assessment (MoCA) scores was observed (B = − 0.045; 95%CI − 0.087, − 0.004; p 0.030). A total of 30 participants were followed-up in 2020. Serum LCN2 levels were decreased in correlation with age (23.31 ± 12.32 ng/ml in 2015 and 15.98 ± 11.28 ng/ml in 2020, p 0.024), while other metabolic parameters were unchanged. Magnetic resonance imaging studies were conducted on a subsample of patients in 2020 (n = 15). Associations between high serum LCN2 levels from 2015 and 2020 and changes in brain volume of hippocampus and prefrontal cortex from 2020 have been observed. These findings suggest a relationship between changes of the level of circulating LCN2, cognitive impairment, and changes in brain volume in patients with MetS. However, further investigation is still needed to explore the direct effect of circulating LCN2 on the cognition of MetS patients.
Collapse
|
33
|
Mann CN, Devi SS, Kersting CT, Bleem AV, Karch CM, Holtzman DM, Gallardo G. Astrocytic α2-Na +/K + ATPase inhibition suppresses astrocyte reactivity and reduces neurodegeneration in a tauopathy mouse model. Sci Transl Med 2022; 14:eabm4107. [PMID: 35171651 DOI: 10.1126/scitranslmed.abm4107] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most dominant form of dementia characterized by the deposition of extracellular amyloid plaques and intracellular neurofibrillary tau tangles (NFTs). In addition to these pathologies, an emerging pathophysiological mechanism that influences AD is neuroinflammation. Astrocytes are a vital type of glial cell that contribute to neuroinflammation, and reactive astrocytes, or astrogliosis, are a well-known pathological feature of AD. However, the mechanisms by which astrocytes contribute to the neurodegenerative process in AD have not been fully elucidated. Here, we showed that astrocytic α2-Na+/K+ adenosine triphosphatase (α2-NKA) is elevated in postmortem human brain tissue from AD and progressive nuclear palsy, a primary tauopathy. The increased astrocytic α2-NKA was also recapitulated in a mouse model of tauopathy. Pharmacological inhibition of α2-NKA robustly suppressed neuroinflammation and reduced brain atrophy. In addition, α2-NKA knockdown in tauopathy mice halted the accumulation of tau pathology. We also demonstrated that α2-NKA promoted tauopathy, in part, by regulating the proinflammatory protein lipocalin-2 (Lcn2). Overexpression of Lcn2 in tauopathy mice increased tau pathology, and prolonged Lcn2 exposure to primary neurons promoted tau uptake in vitro. These studies collectively highlight the contribution of reactive astrocytes to tau pathogenesis in mice and define α2-NKA as a major regulator of astrocytic-dependent neuroinflammation.
Collapse
Affiliation(s)
- Carolyn N Mann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Shamulailatpam Shreedarshanee Devi
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Corey T Kersting
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Amber V Bleem
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Celeste M Karch
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - Gilbert Gallardo
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
34
|
Brattinga B, Plas M, Spikman JM, Rutgers A, de Haan JJ, Absalom AR, van der Wal-Huisman H, de Bock GH, van Leeuwen BL. The association between the inflammatory response following surgery and post-operative delirium in older oncological patients: a prospective cohort study. Age Ageing 2022; 51:afab237. [PMID: 35180288 PMCID: PMC9160877 DOI: 10.1093/ageing/afab237] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/07/2021] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Post-operative delirium (POD) is associated with increased morbidity and mortality rates in older patients. Neuroinflammation, the activation of the intrinsic immune system of the brain, seems to be one of the mechanisms behind the development of POD. The aim of this study was to explore the association between the perioperative inflammatory response and the development of POD in a cohort of older oncological patients in need for surgery. METHODS In this prospective cohort study, patients 65 years and older in need for oncologic surgery were included. Inflammatory markers C-reactive protein (CRP), interleukin-1 beta (IL-1β), IL-6, IL10 and Neutrophil gelatinase-associated lipocalin (NGAL) were measured in plasma samples pre- and post-operatively. Delirium Observation Screening Scale (DOS) was used as screening instrument for POD in the first week after surgery. In case of positive screening, diagnosis of POD was assessed by a clinician. RESULTS Between 2010 and 2016, plasma samples of 311 patients with median age of 72 years (range 65-89) were collected. A total of 38 (12%) patients developed POD in the first week after surgery. The perioperative increase in plasma levels of IL-10 and NGAL were associated with POD in multivariate logistic regression analysis (OR 1.33 [1.09-1.63] P = 0.005 and OR 1.30 [1.03-1.64], P = 0.026, respectively). The biomarkers CRP, IL-1β and IL-6 were not significantly associated with POD. CONCLUSIONS Increased surgery-evoked inflammatory responses of IL-10 and NGAL are associated with the development of POD in older oncological patients. The outcomes of this study contribute to understanding the aetiology of neuroinflammation and the development of POD.
Collapse
Affiliation(s)
- Baukje Brattinga
- University of Groningen, University Medical Center Groningen, Department of Surgery, 9700 RB Groningen, The Netherlands
| | - Matthijs Plas
- University of Groningen, University Medical Center Groningen, Department of Surgery, 9700 RB Groningen, The Netherlands
| | - Jacoba M Spikman
- University of Groningen, University Medical Center Groningen, Department of Neurology, 9700 RB Groningen, The Netherlands
| | - Abraham Rutgers
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, 9700 RB Groningen, The Netherlands
| | - Jacco J de Haan
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, 9700 RB Groningen, The Netherlands
| | - Anthony R Absalom
- University of Groningen, University Medical Center Groningen, Department of Anesthesiology, 9700 RB Groningen, The Netherlands
| | - Hanneke van der Wal-Huisman
- University of Groningen, University Medical Center Groningen, Department of Surgery, 9700 RB Groningen, The Netherlands
| | - Geertruida H de Bock
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, 9700 RB Groningen, The Netherlands
| | - Barbara L van Leeuwen
- University of Groningen, University Medical Center Groningen, Department of Surgery, 9700 RB Groningen, The Netherlands
| |
Collapse
|
35
|
Kim J, Rahman MH, Lee WH, Suk K. Chemogenetic stimulation of the G i pathway in astrocytes suppresses neuroinflammation. Pharmacol Res Perspect 2021; 9:e00822. [PMID: 34676988 PMCID: PMC8532135 DOI: 10.1002/prp2.822] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/30/2021] [Indexed: 11/06/2022] Open
Abstract
Engineered G protein-coupled receptors (GPCRs) are commonly used in chemogenetics as designer receptors exclusively activated by designer drugs (DREADDs). Although several GPCRs have been studied in astrocytes using a chemogenetic approach, the functional role of the astrocytic Gi pathway is not clear, as the literature is conflicting depending on the brain regions or behaviors investigated. In this study, we evaluated the role of the astrocytic Gi pathway in neuroinflammation using a Gi -coupled DREADD (hM4Di). Gi -DREADD was expressed in hippocampal astrocytes of a lipopolysaccharide (LPS)-induced neuroinflammation mouse model using adeno-associated viruses. We found that astrocyte Gi -DREADD stimulation using clozapine N-oxide (CNO) inhibits neuroinflammation, as characterized by decreased levels of proinflammatory cytokines, glial activation, and cognitive impairment in mice. Subsequent experiments using primary astrocyte cultures revealed that Gi -DREADD stimulation significantly downregulated LPS-induced expression of Nos2 mRNA and nitric oxide production. Similarly, in vitro calcium imaging showed that activation of the astrocytic Gi pathway attenuated intracellular calcium transients triggered by LPS treatment, suggesting a positive correlation between enhanced calcium transients and the inflammatory phenotype of astrocytes observed in the inflamed brain. Taken together, our results indicate that the astrocytic Gi pathway plays an inhibitory role in neuroinflammation, providing an opportunity to identify potential cellular and molecular targets to control neuroinflammation.
Collapse
Affiliation(s)
- Jae‐Hong Kim
- Department of PharmacologySchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- BK21 Plus KNU Biomedical Convergence ProgramDepartment of Biomedical SciencesSchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Md Habibur Rahman
- Department of PharmacologySchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- BK21 Plus KNU Biomedical Convergence ProgramDepartment of Biomedical SciencesSchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- Brain Science & Engineering InstituteKyungpook National UniversityDaeguRepublic of Korea
| | - Won Ha Lee
- School of Life SciencesBrain Korea 21 Plus KNU Creative BioResearch GroupKyungpook National UniversityDaeguRepublic of Korea
| | - Kyoungho Suk
- Department of PharmacologySchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- BK21 Plus KNU Biomedical Convergence ProgramDepartment of Biomedical SciencesSchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
- Brain Science & Engineering InstituteKyungpook National UniversityDaeguRepublic of Korea
| |
Collapse
|
36
|
Wang X, Li X, Zuo X, Liang Z, Ding T, Li K, Ma Y, Li P, Zhu Z, Ju C, Zhang Z, Song Z, Quan H, Zhang J, Hu X, Wang Z. Photobiomodulation inhibits the activation of neurotoxic microglia and astrocytes by inhibiting Lcn2/JAK2-STAT3 crosstalk after spinal cord injury in male rats. J Neuroinflammation 2021; 18:256. [PMID: 34740378 PMCID: PMC8571847 DOI: 10.1186/s12974-021-02312-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neurotoxic microglia and astrocytes begin to activate and participate in pathological processes after spinal cord injury (SCI), subsequently causing severe secondary damage and affecting tissue repair. We have previously reported that photobiomodulation (PBM) can promote functional recovery by reducing neuroinflammation after SCI, but little is known about the underlying mechanism. Therefore, we aimed to investigate whether PBM ameliorates neuroinflammation by modulating the activation of microglia and astrocytes after SCI. METHODS Male Sprague-Dawley rats were randomly divided into three groups: a sham control group, an SCI + vehicle group and an SCI + PBM group. PBM was performed for two consecutive weeks after clip-compression SCI models were established. The activation of neurotoxic microglia and astrocytes, the level of tissue apoptosis, the number of motor neurons and the recovery of motor function were evaluated at different days post-injury (1, 3, 7, 14, and 28 days post-injury, dpi). Lipocalin 2 (Lcn2) and Janus kinase-2 (JAK2)-signal transducer and activator of transcription-3 (STAT3) signaling were regarded as potential targets by which PBM affected neurotoxic microglia and astrocytes. In in vitro experiments, primary microglia and astrocytes were irradiated with PBM and cotreated with cucurbitacin I (a JAK2-STAT3 pathway inhibitor), an adenovirus (shRNA-Lcn2) and recombinant Lcn2 protein. RESULTS PBM promoted the recovery of motor function, inhibited the activation of neurotoxic microglia and astrocytes, alleviated neuroinflammation and tissue apoptosis, and increased the number of neurons retained after SCI. The upregulation of Lcn2 and the activation of the JAK2-STAT3 pathway after SCI were suppressed by PBM. In vitro experiments also showed that Lcn2 and JAK2-STAT3 were mutually promoted and that PBM interfered with this interaction, inhibiting the activation of microglia and astrocytes. CONCLUSION Lcn2/JAK2-STAT3 crosstalk is involved in the activation of neurotoxic microglia and astrocytes after SCI, and this process can be suppressed by PBM.
Collapse
Affiliation(s)
- Xuankang Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xin Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.,967 Hospital of People's Liberation Army Joint Logistic Support Force, Dalian, 116044, Liaoning, China
| | - Xiaoshuang Zuo
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhuowen Liang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Tan Ding
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Kun Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yangguang Ma
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Penghui Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhijie Zhu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Cheng Ju
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhihao Zhang
- 967 Hospital of People's Liberation Army Joint Logistic Support Force, Dalian, 116044, Liaoning, China
| | - Zhiwen Song
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Huilin Quan
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jiawei Zhang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xueyu Hu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Zhe Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
37
|
Role of Lipocalin-2 in Amyloid-Beta Oligomer-Induced Mouse Model of Alzheimer's Disease. Antioxidants (Basel) 2021; 10:antiox10111657. [PMID: 34829528 PMCID: PMC8614967 DOI: 10.3390/antiox10111657] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
Lipocalin-2 (LCN2) is an inflammatory protein with diverse functions in the brain. Although many studies have investigated the mechanism of LCN2 in brain injuries, the effect of LCN2 on amyloid-toxicity-related memory deficits in a mouse model of Alzheimer’s disease (AD) has been less studied. We investigated the role of LCN2 in human AD patients using a mouse model of AD. We created an AD mouse model by injecting amyloid-beta oligomer (AβO) into the hippocampus. In this model, animals exhibited impaired learning and memory. We found LCN2 upregulation in the human brain frontal lobe, as well as a positive correlation between white matter ischemic changes and serum LCN2. We also found increased astrocytic LCN2, microglia activation, iron accumulation, and blood–brain barrier disruption in AβO-treated hippocampi. These findings suggest that LCN2 is involved in a variety of amyloid toxicity mechanisms, especially neuroinflammation and oxidative stress.
Collapse
|
38
|
Olson B, Norgard MA, Levasseur PR, Zhu X, Marks DL. Physiologic and molecular characterization of a novel murine model of metastatic head and neck cancer cachexia. J Cachexia Sarcopenia Muscle 2021; 12:1312-1332. [PMID: 34231343 PMCID: PMC8517353 DOI: 10.1002/jcsm.12745] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/19/2021] [Accepted: 06/08/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cancer cachexia is a metabolic disorder characterized by the progressive loss of fat and lean mass that results in significant wasting, ultimately leading to reduced quality of life and increased mortality. Effective therapies for cachexia are lacking, potentially owing to the mismatch in clinically relevant models of cachexia. Specifically, cachexia observed in a clinical setting is commonly associated with advanced or late-stage cancers that are metastatic, yet pre-clinical metastatic models of cachexia are limited. Furthermore, the prevalence of cachexia in head and neck cancer patients is high, yet few pre-clinical models of head and neck cancer cachexia exist. In addition to these shortcomings, cachexia is also heterogeneous among any given cancer, whereas patients with similar disease burden may experience significantly different degrees of cachexia symptoms. In order to address these issues, we characterize a metastatic model of human papilloma virus (HPV) positive head and neck squamous cell carcinoma that recapitulates the cardinal clinical and molecular features of cancer cachexia. METHODS Male and female C57BL/6 mice were implanted subcutaneously with oropharyngeal squamous cell carcinoma cells stably transformed with HPV16 E6 and E7 together with hRas and luciferase (mEERL) that metastasizes to the lungs (MLM). We then robustly characterize the physiologic, behavioural, and molecular signatures during tumour development in two MLM subclones. RESULTS Mice injected with MLM tumour cells rapidly developed primary tumours and eventual metastatic lesions to the lungs. MLM3, but not MLM5, engrafted mice progressively lost fat and lean mass during tumour development despite the absence of anorexia (P < 0.05). Behaviourally, MLM3-implanted mice displayed decreased locomotor behaviours and impaired nest building (P < 0.05). Muscle catabolism programmes associated with cachexia, including E3 ubiquitin ligase and autophagy up-regulation, along with progressive adipose wasting and accompanying browning gene signatures, were observed. Tumour progression also corresponded with hypothalamic and peripheral organ inflammation, as well as an elevation in neutrophil-to-lymphocyte ratio (P < 0.05). Finally, we characterize the fat and lean mass sparing effects of voluntary wheel running on MLM3 cachexia (P < 0.05). CONCLUSIONS This syngeneic MLM3 allograft model of metastatic cancer cachexia is reliable, consistent, and readily recapitulates key clinical and molecular features and heterogeneity of cancer cachexia. Because few metastatic models of cachexia exist-even though cachexia often accompanies metastatic progression-we believe this model more accurately captures cancer cachexia observed in a clinical setting and thus is well suited for future mechanistic studies and pre-clinical therapy development for this crippling metabolic disorder.
Collapse
Affiliation(s)
- Brennan Olson
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandORUSA
- Medical Scientist Training ProgramOregon Health & Science UniversityPortlandORUSA
| | - Mason A. Norgard
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandORUSA
| | - Peter R. Levasseur
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandORUSA
| | - Xinxia Zhu
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandORUSA
| | - Daniel L. Marks
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandORUSA
- Brenden‐Colson Center for Pancreatic CareOregon Health and & Science University PortlandORUSA
- Knight Cancer InstituteOregon Health & Science UniversityPortlandORUSA
| |
Collapse
|
39
|
Cyanotoxins and the Nervous System. Toxins (Basel) 2021; 13:toxins13090660. [PMID: 34564664 PMCID: PMC8472772 DOI: 10.3390/toxins13090660] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/31/2021] [Accepted: 09/09/2021] [Indexed: 11/16/2022] Open
Abstract
Cyanobacteria are capable of producing a wide range of bioactive compounds with many considered to be toxins. Although there are a number of toxicological outcomes with respect to cyanobacterial exposure, this review aims to examine those which affect the central nervous system (CNS) or have neurotoxicological properties. Such exposures can be acute or chronic, and we detail issues concerning CNS entry, detection and remediation. Exposure can occur through a variety of media but, increasingly, exposure through air via inhalation may have greater significance and requires further investigation. Even though cyanobacterial toxins have traditionally been classified based on their primary mode of toxicity, increasing evidence suggests that some also possess neurotoxic properties and include known cyanotoxins and unknown compounds. Furthermore, chronic long-term exposure to these compounds is increasingly being identified as adversely affecting human health.
Collapse
|
40
|
Dekens DW, Eisel ULM, Gouweleeuw L, Schoemaker RG, De Deyn PP, Naudé PJW. Lipocalin 2 as a link between ageing, risk factor conditions and age-related brain diseases. Ageing Res Rev 2021; 70:101414. [PMID: 34325073 DOI: 10.1016/j.arr.2021.101414] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022]
Abstract
Chronic (neuro)inflammation plays an important role in many age-related central nervous system (CNS) diseases, including Alzheimer's disease, Parkinson's disease and vascular dementia. Inflammation also characterizes many conditions that form a risk factor for these CNS disorders, such as physical inactivity, obesity and cardiovascular disease. Lipocalin 2 (Lcn2) is an inflammatory protein shown to be involved in different age-related CNS diseases, as well as risk factor conditions thereof. Lcn2 expression is increased in the periphery and the brain in different age-related CNS diseases and also their risk factor conditions. Experimental studies indicate that Lcn2 contributes to various neuropathophysiological processes of age-related CNS diseases, including exacerbated neuroinflammation, cell death and iron dysregulation, which may negatively impact cognitive function. We hypothesize that increased Lcn2 levels as a result of age-related risk factor conditions may sensitize the brain and increase the risk to develop age-related CNS diseases. In this review we first provide a comprehensive overview of the known functions of Lcn2, and its effects in the CNS. Subsequently, this review explores Lcn2 as a potential (neuro)inflammatory link between different risk factor conditions and the development of age-related CNS disorders. Altogether, evidence convincingly indicates Lcn2 as a key constituent in ageing and age-related brain diseases.
Collapse
Affiliation(s)
- Doortje W Dekens
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Leonie Gouweleeuw
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Regien G Schoemaker
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Peter P De Deyn
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Laboratory of Neurochemistry and Behaviour, Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Petrus J W Naudé
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands; Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
41
|
Lipocalin 2 as a Putative Modulator of Local Inflammatory Processes in the Spinal Cord and Component of Organ Cross talk After Spinal Cord Injury. Mol Neurobiol 2021; 58:5907-5919. [PMID: 34417948 DOI: 10.1007/s12035-021-02530-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
Lipocalin 2 (LCN2), an immunomodulator, regulates various cellular processes such as iron transport and defense against bacterial infection. Under pathological conditions, LCN2 promotes neuroinflammation via the recruitment and activation of immune cells and glia, particularly microglia and astrocytes. Although it seems to have a negative influence on the functional outcome in spinal cord injury (SCI), the extent of its involvement in SCI and the underlying mechanisms are not yet fully known. In this study, using a SCI contusion mouse model, we first investigated the expression pattern of Lcn2 in different parts of the CNS (spinal cord and brain) and in the liver and its concentration in blood serum. Interestingly, we could note a significant increase in LCN2 throughout the whole spinal cord, in the brain, liver, and blood serum. This demonstrates the diversity of its possible sites of action in SCI. Furthermore, genetic deficiency of Lcn2 (Lcn2-/-) significantly reduced certain aspects of gliosis in the SCI-mice. Taken together, our studies provide first valuable hints, suggesting that LCN2 is involved in the local and systemic effects post SCI, and might modulate the impairment of different peripheral organs after injury.
Collapse
|
42
|
Weng YC, Huang YT, Chiang IC, Tsai PJ, Su YW, Chou WH. Lipocalin-2 mediates the rejection of neural transplants. FASEB J 2021; 35:e21317. [PMID: 33421207 DOI: 10.1096/fj.202001018r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 11/19/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Lipocalin-2 (LCN2) has been implicated in promoting apoptosis and neuroinflammation in neurological disorders; however, its role in neural transplantation remains unknown. In this study, we cultured and differentiated Lund human mesencephalic (LUHMES) cells into human dopaminergic-like neurons and found that LCN2 mRNA was progressively induced in mouse brain after the intrastriatal transplantation of human dopaminergic-like neurons. The induction of LCN2 protein was detected in a subset of astrocytes and neutrophils infiltrating the core of the engrafted sites, but not in neurons and microglia. LCN2-immunoreactive astrocytes within the engrafted sites expressed lower levels of A1 and A2 astrocytic markers. Recruitment of microglia, neutrophils, and monocytes after transplantation was attenuated in LCN2 deficiency mice. The expression of M2 microglial markers was significantly elevated and survival of engrafted neurons was markedly improved after transplantation in LCN2 deficiency mice. Brain type organic cation transporter (BOCT), the cell surface receptor for LCN2, was induced in dopaminergic-like neurons after differentiation, and treatment with recombinant LCN2 protein directly induced apoptosis in dopaminergic-like neurons in a dose-dependent manner. Our results, therefore, suggested that LCN2 is a neurotoxic factor for the engrafted neurons and a modulator of neuroinflammation. LCN2 inhibition may be useful in reducing rejection after neural transplantation.
Collapse
Affiliation(s)
- Yi-Chinn Weng
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Ting Huang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - I-Chen Chiang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Ju Tsai
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Wen-Hai Chou
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
43
|
das Neves SP, Taipa R, Marques F, Soares Costa P, Monárrez-Espino J, Palha JA, Kivipelto M. Association Between Iron-Related Protein Lipocalin 2 and Cognitive Impairment in Cerebrospinal Fluid and Serum. Front Aging Neurosci 2021; 13:663837. [PMID: 34248600 PMCID: PMC8267056 DOI: 10.3389/fnagi.2021.663837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/30/2021] [Indexed: 11/24/2022] Open
Abstract
A worldwide increase in longevity is bringing novel challenges to public health and health care professionals. Cognitive impairment in the elderly may compromise living conditions and precede Alzheimer’s disease (AD), the most prevalent form of dementia. Therefore, finding molecular markers associated with cognitive impairment is of crucial importance. Lipocalin 2 (LCN2), an iron-related protein, has been suggested as a potential marker for mild cognitive impairment (MCI) and AD. This study aimed at investigating the association between LCN2 measured in serum and cerebrospinal fluid (CSF) with cognitive impairment. A cross-sectional design based on two aging cohorts was used: individuals diagnosed with subjective cognitive complaints (SCC), MCI, and AD from a Swedish memory clinic-based cohort, and individuals diagnosed with SCC and AD from a Portuguese cohort. Binary logistic [for the outcome cognitive impairment (MCI + AD) in the Swedish cohort and AD in the Portuguese cohort] and multinomial logistic (for the outcomes MCI and AD) regression analyses were used. No associations were found in both cohorts when controlling for sex, education, and age. This explanatory study suggests that the association between serum and CSF LCN2 concentrations with cognitive impairment reported in the literature must be further analyzed for confounders.
Collapse
Affiliation(s)
- Sofia Pereira das Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo Taipa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Department of Neurosciences, Centro Hospitalar do Porto, Porto, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrício Soares Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joel Monárrez-Espino
- Department of Health Research, Christus Muguerza Hospital-University of Monterrey, Chihuahua, Mexico
| | - Joana A Palha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
44
|
Chen H, Shang D, Wen Y, Liang C. Bone-Derived Modulators That Regulate Brain Function: Emerging Therapeutic Targets for Neurological Disorders. Front Cell Dev Biol 2021; 9:683457. [PMID: 34179014 PMCID: PMC8222721 DOI: 10.3389/fcell.2021.683457] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/18/2021] [Indexed: 12/31/2022] Open
Abstract
Bone has traditionally been regarded as a structural organ that supports and protects the various organs of the body. Recent studies suggest that bone also acts as an endocrine organ to regulate whole-body metabolism. Particularly, homeostasis of the bone is shown to be necessary for brain development and function. Abnormal bone metabolism is associated with the onset and progression of neurological disorders. Recently, multiple bone-derived modulators have been shown to participate in brain function and neurological disorders, including osteocalcin, lipocalin 2, and osteopontin, as have bone marrow-derived cells such as mesenchymal stem cells, hematopoietic stem cells, and microglia-like cells. This review summarizes current findings regarding the roles of these bone-derived modulators in the brain, and also follows their involvement in the pathogenesis of neurological disorders. The content of this review may aide in the development of promising therapeutic strategies for neurological disorders via targeting bone.
Collapse
Affiliation(s)
- Hongzhen Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, China.,Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dewei Shang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuguan Wen
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
45
|
Zarfeshani A, Carroll KR, Volpe BT, Diamond B. Cognitive Impairment in SLE: Mechanisms and Therapeutic Approaches. Curr Rheumatol Rep 2021; 23:25. [PMID: 33782842 PMCID: PMC11207197 DOI: 10.1007/s11926-021-00992-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
A wide range of patients with systemic lupus erythematosus (SLE) suffer from cognitive dysfunction (CD) which severely impacts their quality of life. However, CD remains underdiagnosed and poorly understood. Here, we discuss current findings in patients and in animal models. Strong evidence suggests that CD pathogenesis involves known mechanisms of tissue injury in SLE. These mechanisms recruit brain resident cells, in particular microglia, into the pathological process. While systemic immune activation is critical to central nervous system injury, the current focus of therapy is the microglial cell and not the systemic immune perturbation. Further studies are critical to examine additional potential therapeutic targets and more specific treatments based on the cause and progress of the disease.
Collapse
Affiliation(s)
- Aida Zarfeshani
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kaitlin R Carroll
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Bruce T Volpe
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
46
|
Bhusal A, Lee WH, Suk K. Lipocalin-2 in Diabetic Complications of the Nervous System: Physiology, Pathology, and Beyond. Front Physiol 2021; 12:638112. [PMID: 33613327 PMCID: PMC7892766 DOI: 10.3389/fphys.2021.638112] [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: 12/05/2020] [Accepted: 01/19/2021] [Indexed: 01/04/2023] Open
Abstract
Lipocalin-2 (LCN2) is a 25 kDa secreted protein that belongs to the family of lipocalins, a group of transporters of small hydrophobic molecules such as iron, fatty acids, steroids, and lipopolysaccharide in circulation. LCN2 was previously found to be involved in iron delivery, pointing toward a potential role for LCN2 in immunity. This idea was further validated when LCN2 was found to limit bacterial growth during infections in mice by sequestering iron-laden siderophores. Recently, LCN2 was also identified as a critical regulator of energy metabolism, glucose and lipid homeostasis, and insulin function. Furthermore, studies using Lcn2 knockout mice suggest an important role for LCN2 in several biobehavioral responses, including cognition, emotion, anxiety, and feeding behavior. Owing to its expression and influence on multiple metabolic and neurological functions, there has emerged a great deal of interest in the study of relationships between LCN2 and neurometabolic complications. Thorough investigation has demonstrated that LCN2 is involved in several neurodegenerative diseases, while more recent studies have shown that LCN2 is also instrumental for the progression of diabetic complications like encephalopathy and peripheral neuropathy. Preliminary findings have shown that LCN2 is also a promising drug target and diagnostic marker for the treatment of neuropathic complications from diabetes. In particular, future translational research related to LCN2, such as the development of small-molecule inhibitors or neutralizing antibodies against LCN2, appears essential for exploring its potential as a therapeutic target.
Collapse
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, South Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, South Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
47
|
Gachpazan M, Akhlaghipour I, Rahimi HR, Saburi E, Mojarrad M, Abbaszadegan MR, Moghbeli M. Genetic and molecular biology of systemic lupus erythematosus among Iranian patients: an overview. AUTO- IMMUNITY HIGHLIGHTS 2021; 12:2. [PMID: 33516274 PMCID: PMC7847600 DOI: 10.1186/s13317-020-00144-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a clinicopathologically heterogeneous chronic autoimmune disorder affecting different organs and tissues. It has been reported that there is an increasing rate of SLE incidence among Iranian population. Moreover, the Iranian SLE patients have more severe clinical manifestations compared with other countries. Therefore, it is required to introduce novel methods for the early detection of SLE in this population. Various environmental and genetic factors are involved in SLE progression. MAIN BODY In present review we have summarized all of the reported genes which have been associated with clinicopathological features of SLE among Iranian patients. CONCLUSIONS Apart from the reported cytokines and chemokines, it was interestingly observed that the apoptosis related genes and non-coding RNAs were the most reported genetic abnormalities associated with SLE progression among Iranians. This review clarifies the genetics and molecular biology of SLE progression among Iranian cases. Moreover, this review paves the way of introducing an efficient panel of genetic markers for the early detection and better management of SLE in this population.
Collapse
Affiliation(s)
- Meisam Gachpazan
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
48
|
Lipocalin-2 Deficiency Reduces Oxidative Stress and Neuroinflammation and Results in Attenuation of Kainic Acid-Induced Hippocampal Cell Death. Antioxidants (Basel) 2021; 10:antiox10010100. [PMID: 33445746 PMCID: PMC7828212 DOI: 10.3390/antiox10010100] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/30/2020] [Accepted: 01/11/2021] [Indexed: 01/03/2023] Open
Abstract
The hippocampal cell death that follows kainic acid (KA)-induced seizures is associated with blood–brain barrier (BBB) leakage and oxidative stress. Lipocalin-2 (LCN2) is an iron-trafficking protein which contributes to both oxidative stress and inflammation. However, LCN2′s role in KA-induced hippocampal cell death is not clear. Here, we examine the effect of blocking LCN2 genetically on neuroinflammation and oxidative stress in KA-induced neuronal death. LCN2 deficiency reduced neuronal cell death and BBB leakage in the KA-treated hippocampus. In addition to LCN2 upregulation in the KA-treated hippocampus, circulating LCN2 levels were significantly increased in KA-treated wild-type (WT) mice. In LCN2 knockout mice, we found that the expressions of neutrophil markers myeloperoxidase and neutrophil elastase were decreased compared to their expressions in WT mice following KA treatment. Furthermore, LCN2 deficiency also attenuated KA-induced iron overload and oxidative stress in the hippocampus. These findings indicate that LCN2 may play an important role in iron-related oxidative stress and neuroinflammation in KA-induced hippocampal cell death.
Collapse
|
49
|
Bhusal A, Rahman MH, Lee WH, Lee IK, Suk K. Satellite glia as a critical component of diabetic neuropathy: Role of lipocalin-2 and pyruvate dehydrogenase kinase-2 axis in the dorsal root ganglion. Glia 2020; 69:971-996. [PMID: 33251681 DOI: 10.1002/glia.23942] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Diabetic peripheral neuropathy (DPN) is a common complication of uncontrolled diabetes. The pathogenesis of DPN is associated with chronic inflammation in dorsal root ganglion (DRG), eventually causing structural and functional changes. Studies on DPN have primarily focused on neuronal component, and there is limited knowledge about the role of satellite glial cells (SGCs), although they completely enclose neuronal soma in DRG. Lipocalin-2 (LCN2) is a pro-inflammatory acute-phase protein found in high levels in diverse neuroinflammatory and metabolic disorders. In diabetic DRG, the expression of LCN2 was increased exclusively in the SGCs. This upregulation of LCN2 in SGCs correlated with increased inflammatory responses in DRG and sciatic nerve. Furthermore, diabetes-induced inflammation and morphological changes in DRG, as well as sciatic nerve, were attenuated in Lcn2 knockout (KO) mice. Lcn2 gene ablation also ameliorated neuropathy phenotype as determined by nerve conduction velocity and intraepidermal nerve fiber density. Mechanistically, studies using specific gene KO mice, adenovirus-mediated gene overexpression strategy, and primary cultures of DRG SGCs and neurons have demonstrated that LCN2 enhances the expression of mitochondrial gate-keeping regulator pyruvate dehydrogenase kinase-2 (PDK2) through PPARβ/δ, thereby inhibiting pyruvate dehydrogenase activity and increasing production of glycolytic end product lactic acid in DRG SGCs and neurons of diabetic mice. Collectively, our findings reveal a crucial role of glial LCN2-PPARβ/δ-PDK2-lactic acid axis in progression of DPN. Our results establish a link between pro-inflammatory LCN2 and glycolytic PDK2 in DRG SGCs and neurons and propose a novel glia-based mechanism and drug target for therapy of DPN. MAIN POINTS: Diabetes upregulates LCN2 in satellite glia, which in turn increases pyruvate dehydrogenase kinase-2 (PDK2) expression and lactic acid production in dorsal root ganglia (DRG). Glial LCN2-PDK2-lactic acid axis in DRG plays a crucial role in the pathogenesis of diabetic neuropathy.
Collapse
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, Brain Korea 21 Plus/Kyungpook National University Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.,Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
50
|
Czapski GA, Zhao Y, Lukiw WJ, Strosznajder JB. Acute Systemic Inflammatory Response Alters Transcription Profile of Genes Related to Immune Response and Ca 2+ Homeostasis in Hippocampus; Relevance to Neurodegenerative Disorders. Int J Mol Sci 2020; 21:ijms21217838. [PMID: 33105802 PMCID: PMC7660108 DOI: 10.3390/ijms21217838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 12/22/2022] Open
Abstract
Acute systemic inflammatory response (SIR) triggers an alteration in the transcription of brain genes related to neuroinflammation, oxidative stress and cells death. These changes are also characteristic for Alzheimer’s disease (AD) neuropathology. Our aim was to evaluate gene expression patterns in the mouse hippocampus (MH) by using microarray technology 12 and 96 h after SIR evoked by lipopolysaccharide (LPS). The results were compared with microarray analysis of human postmortem hippocampal AD tissues. It was found that 12 h after LPS administration the expression of 231 genes in MH was significantly altered (FC > 2.0); however, after 96 h only the S100a8 gene encoding calgranulin A was activated (FC = 2.9). Gene ontology enrichment analysis demonstrated the alteration of gene expression related mostly to the immune-response including the gene Lcn2 for Lipocalin 2 (FC = 237.8), involved in glia neurotoxicity. The expression of genes coding proteins involved in epigenetic regulation, histone deacetylases (Hdac4,5,8,9,11) and bromo- and extraterminal domain protein Brd3 were downregulated; however, Brd2 was found to be upregulated. Remarkably, the significant increase in expression of Lcn2, S100a8, S100a9 and also Saa3 and Ch25h, was found in AD brains suggesting that early changes of immune-response genes evoked by mild SIR could be crucial in AD pathogenesis.
Collapse
Affiliation(s)
- Grzegorz A. Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
- Correspondence: (G.A.C.); (J.B.S.); Tel.: +48-22-6086-600 (G.A.C.); +48-22-6086-414 (J.B.S.)
| | - Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Science Center (LSU-HSC), New Orleans, LA 70112, USA; (Y.Z.); (W.J.L.)
- Department of Cell Biology and Anatomy, LSU-HSC, New Orleans, LA 70112, USA
| | - Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center (LSU-HSC), New Orleans, LA 70112, USA; (Y.Z.); (W.J.L.)
- Department of Ophthalmology, LSU-HSC, New Orleans, LA 70112, USA
- Department of Neurology, LSU-HSC, New Orleans, LA 70112, USA
| | - Joanna B. Strosznajder
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
- Correspondence: (G.A.C.); (J.B.S.); Tel.: +48-22-6086-600 (G.A.C.); +48-22-6086-414 (J.B.S.)
| |
Collapse
|