1
|
Lin Z, Ying C, Si X, Xue N, Liu Y, Zheng R, Chen Y, Pu J, Zhang B. NOX4 exacerbates Parkinson's disease pathology by promoting neuronal ferroptosis and neuroinflammation. Neural Regen Res 2025; 20:2038-2052. [PMID: 38993139 DOI: 10.4103/nrr.nrr-d-23-01265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 03/18/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00026/figure1/v/2024-09-09T124005Z/r/image-tiff Parkinson's disease is primarily caused by the loss of dopaminergic neurons in the substantia nigra compacta. Ferroptosis, a novel form of regulated cell death characterized by iron accumulation and lipid peroxidation, plays a vital role in the death of dopaminergic neurons. However, the molecular mechanisms underlying ferroptosis in dopaminergic neurons have not yet been completely elucidated. NADPH oxidase 4 is related to oxidative stress, however, whether it regulates dopaminergic neuronal ferroptosis remains unknown. The aim of this study was to determine whether NADPH oxidase 4 is involved in dopaminergic neuronal ferroptosis, and if so, by what mechanism. We found that the transcriptional regulator activating transcription factor 3 increased NADPH oxidase 4 expression in dopaminergic neurons and astrocytes in an 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine-induced Parkinson's disease model. NADPH oxidase 4 inhibition improved the behavioral impairments observed in the Parkinson's disease model animals and reduced the death of dopaminergic neurons. Moreover, NADPH oxidase 4 inhibition reduced lipid peroxidation and iron accumulation in the substantia nigra of the Parkinson's disease model animals. Mechanistically, we found that NADPH oxidase 4 interacted with activated protein kinase C α to prevent ferroptosis of dopaminergic neurons. Furthermore, by lowering the astrocytic lipocalin-2 expression, NADPH oxidase 4 inhibition reduced 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine-induced neuroinflammation. These findings demonstrate that NADPH oxidase 4 promotes ferroptosis of dopaminergic neurons and neuroinflammation, which contribute to dopaminergic neuron death, suggesting that NADPH oxidase 4 is a possible therapeutic target for Parkinson's disease.
Collapse
Affiliation(s)
- Zhihao Lin
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Si W, You R, Sun B, Luo J, Hu G. The role of LCN2 in exacerbating ferroptosis levels in acute ischemic stroke injury. Biochem Biophys Res Commun 2024; 733:150452. [PMID: 39067246 DOI: 10.1016/j.bbrc.2024.150452] [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: 07/20/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Due to the complex pathogenesis of acute ischemic stroke (AIS), further investigation into its underlying mechanisms is necessary. Presently, existing literature indicates a close association between ferroptosis and AIS injury; however, the precise mechanism and molecular target of ferroptosis in AIS injury remain elusive. By RNA sequencing, we found a significant increase in LCN2 expression in the ischemic cortex. In order to investigate the potential role of LCN2 in modulating AIS injury through the regulation of ferroptosis, we utilized RNA interference (RNAi) knockdown and gene overexpression experiments. The findings from experiments conducted both in vitro and in vivo revealed a marked increase in ferroptosis levels within the AIS model group. Suppression of the LCN2 gene resulted in a significant reduction in ferroptosis levels in OGD/R cells. Conversely, upregulation of LCN2 exacerbated ferroptosis levels in OGD/R cells. The results suggest that elevated levels of ferroptosis may result from heightened expression of LCN2, thereby exacerbating ischemia/reperfusion injury. This study indicates the involvement of ferroptosis in the pathogenesis of AIS and highlights LCN2 as a regulator of ferroptosis in AIS-induced injury, suggesting a potential therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Wenwen Si
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, 518104, PR China.
| | - Ruijia You
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, PR China
| | - Bin Sun
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, PR China
| | - Jing Luo
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, PR China
| | - Guanhua Hu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, PR China
| |
Collapse
|
3
|
Zubareva OE, Kharisova AR, Roginskaya AI, Kovalenko AA, Zakharova MV, Schwarz AP, Sinyak DS, Zaitsev AV. PPARβ/δ Agonist GW0742 Modulates Microglial and Astroglial Gene Expression in a Rat Model of Temporal Lobe Epilepsy. Int J Mol Sci 2024; 25:10015. [PMID: 39337503 PMCID: PMC11432388 DOI: 10.3390/ijms251810015] [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: 08/14/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
The role of astroglial and microglial cells in the pathogenesis of epilepsy is currently under active investigation. It has been proposed that the activity of these cells may be regulated by the agonists of peroxisome proliferator-activated nuclear receptors (PPARs). This study investigated the effects of a seven-day treatment with the PPAR β/δ agonist GW0742 (Fitorine, 5 mg/kg/day) on the behavior and gene expression of the astroglial and microglial proteins involved in the regulation of epileptogenesis in the rat brain within a lithium-pilocarpine model of temporal lobe epilepsy (TLE). TLE resulted in decreased social and increased locomotor activity in the rats, increased expression of astro- and microglial activation marker genes (Gfap, Aif1), pro- and anti-inflammatory cytokine genes (Tnfa, Il1b, Il1rn), and altered expression of other microglial (Nlrp3, Arg1) and astroglial (Lcn2, S100a10) genes in the dorsal hippocampus and cerebral cortex. GW0742 attenuated, but did not completely block, some of these impairments. Specifically, the treatment affected Gfap gene expression in the dorsal hippocampus and Aif1 gene expression in the cortex. The GW0742 injections attenuated the TLE-specific enhancement of Nlrp3 and Il1rn gene expression in the cortex. These results suggest that GW0742 may affect the expression of some genes involved in the regulation of epileptogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Aleksey V. Zaitsev
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 194223 Saint Petersburg, Russia; (O.E.Z.); (A.R.K.); (A.I.R.); (A.A.K.); (M.V.Z.); (A.P.S.); (D.S.S.)
| |
Collapse
|
4
|
Xiao Y, Wang G, He G, Qin W, Shi Y. Rab8a/SNARE complex activation promotes vesicle anchoring and transport in spinal astrocytes to drive neuropathic pain. BIOMOLECULES & BIOMEDICINE 2024; 24:1290-1300. [PMID: 38691557 PMCID: PMC11379012 DOI: 10.17305/bb.2024.10441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
Neuropathic pain (NPP) remains a clinically challenging condition, driven by the activation of spinal astrocytes and the complex release of inflammatory mediators. This study aimed to examine the roles of Rab8a and SNARE complex proteins in activated astrocytes to uncover the underlying mechanisms of NPP. The research was conducted using a rat model with chronic constriction injury (CCI) of the sciatic nerve and primary astrocytes treated with lipopolysaccharide. Enhanced expression of Rab8a was noted specifically in spinal dorsal horn astrocytes through immunofluorescence. Electron microscopy observations showed increased vesicular transport and exocytic activity in activated astrocytes, which was corroborated by elevated levels of inflammatory cytokines such as interleukin (IL)-1β and tumor necrosis factor (TNF)-α detected through quantitative PCR. Western blot analyses confirmed significant upregulation of Rab8a, VAMP2, and Syntaxin16 in these cells. Furthermore, the application of botulinum neurotoxin type A (BONT/A) reduced the levels of vesicle transport-associated proteins, inhibiting vesicular transport in activated astrocytes. These findings suggest that the Rab8a/SNARE pathway in astrocytes enhances vesicle transport and anchoring, increasing the secretion of bioactive molecules that may play a crucial role in the pathophysiology of NPP. Inhibiting this pathway with BONT/A offers a novel therapeutic target for managing NPP, highlighting its potential utility in clinical interventions.
Collapse
Affiliation(s)
- Yunqiao Xiao
- University-Town Hospital of Chongqing Medical University, Chongqing, China
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Gengyi Wang
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Guiqiong He
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Wanxiang Qin
- Department of Pain Care, Southwest Hospital, Army Medical University, Chongqing, China
| | - Ying Shi
- Department of Pain Care, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
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
|
6
|
Xu H, Li H, Zhang P, Gao Y, Ma H, Gao T, Liu H, Hua W, Zhang L, Zhang X, Yang P, Liu J. The functions of exosomes targeting astrocytes and astrocyte-derived exosomes targeting other cell types. Neural Regen Res 2024; 19:1947-1953. [PMID: 38227520 PMCID: PMC11040311 DOI: 10.4103/1673-5374.390961] [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: 05/27/2023] [Revised: 07/20/2023] [Accepted: 09/08/2023] [Indexed: 01/17/2024] Open
Abstract
Astrocytes are the most abundant glial cells in the central nervous system; they participate in crucial biological processes, maintain brain structure, and regulate nervous system function. Exosomes are cell-derived extracellular vesicles containing various bioactive molecules including proteins, peptides, nucleotides, and lipids secreted from their cellular sources. Increasing evidence shows that exosomes participate in a communication network in the nervous system, in which astrocyte-derived exosomes play important roles. In this review, we have summarized the effects of exosomes targeting astrocytes and the astrocyte-derived exosomes targeting other cell types in the central nervous system. We also discuss the potential research directions of the exosome-based communication network in the nervous system. The exosome-based intercellular communication focused on astrocytes is of great significance to the biological and/or pathological processes in different conditions in the brain. New strategies may be developed for the diagnosis and treatment of neurological disorders by focusing on astrocytes as the central cells and utilizing exosomes as communication mediators.
Collapse
Affiliation(s)
- Hongye Xu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - He Li
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Emergency, Naval Hospital of Eastern Theater, Zhoushan, Zhejiang Province, China
| | - Ping Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yuan Gao
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hongyu Ma
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tianxiang Gao
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hanchen Liu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Weilong Hua
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lei Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaoxi Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Pengfei Yang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianmin Liu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
7
|
Boonpraman N, Yi SS. NADPH oxidase 4 (NOX4) as a biomarker and therapeutic target in neurodegenerative diseases. Neural Regen Res 2024; 19:1961-1966. [PMID: 38227522 DOI: 10.4103/1673-5374.390973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/25/2023] [Indexed: 01/17/2024] Open
Abstract
Diseases like Alzheimer's and Parkinson's diseases are defined by inflammation and the damage neurons undergo due to oxidative stress. A primary reactive oxygen species contributor in the central nervous system, NADPH oxidase 4, is viewed as a potential therapeutic touchstone and indicative marker for these ailments. This in-depth review brings to light distinct features of NADPH oxidase 4, responsible for generating superoxide and hydrogen peroxide, emphasizing its pivotal role in activating glial cells, inciting inflammation, and disturbing neuronal functions. Significantly, malfunctioning astrocytes, forming the majority in the central nervous system, play a part in advancing neurodegenerative diseases, due to their reactive oxygen species and inflammatory factor secretion. Our study reveals that aiming at NADPH oxidase 4 within astrocytes could be a viable treatment pathway to reduce oxidative damage and halt neurodegenerative processes. Adjusting NADPH oxidase 4 activity might influence the neuroinflammatory cytokine levels, including myeloperoxidase and osteopontin, offering better prospects for conditions like Alzheimer's disease and Parkinson's disease. This review sheds light on the role of NADPH oxidase 4 in neural degeneration, emphasizing its drug target potential, and paving the path for novel treatment approaches to combat these severe conditions.
Collapse
Affiliation(s)
- Napissara Boonpraman
- BK21 four Program, Department of Medical Sciences, Soonchunhyang University, Asan, South Korea
| | - Sun Shin Yi
- BK21 four Program, Department of Medical Sciences, Soonchunhyang University, Asan, South Korea
- Department of Biomedical Laboratory Science, Soonchunhyang University, Asan, South Korea
- iConnectome, Co., Ltd., Cheonan, South Korea
| |
Collapse
|
8
|
Barnett D, Zimmer TS, Booraem C, Palaguachi F, Meadows SM, Xiao H, Chouchani ET, Orr AG, Orr AL. Mitochondrial complex III-derived ROS amplify immunometabolic changes in astrocytes and promote dementia pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608708. [PMID: 39229090 PMCID: PMC11370371 DOI: 10.1101/2024.08.19.608708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Neurodegenerative disorders alter mitochondrial functions, including the production of reactive oxygen species (ROS). Mitochondrial complex III (CIII) generates ROS implicated in redox signaling, but its triggers, targets, and disease relevance are not clear. Using site-selective suppressors and genetic manipulations together with mitochondrial ROS imaging and multiomic profiling, we found that CIII is the dominant source of ROS production in astrocytes exposed to neuropathology-related stimuli. Astrocytic CIII-ROS production was dependent on nuclear factor-κB (NF-κB) and the mitochondrial sodium-calcium exchanger (NCLX) and caused oxidation of select cysteines within immune and metabolism-associated proteins linked to neurological disease. CIII-ROS amplified metabolomic and pathology-associated transcriptional changes in astrocytes, with STAT3 activity as a major mediator, and facilitated neuronal toxicity in a non-cell-autonomous manner. As proof-of-concept, suppression of CIII-ROS in mice decreased dementia-linked tauopathy and neuroimmune cascades and extended lifespan. Our findings establish CIII-ROS as an important immunometabolic signal transducer and tractable therapeutic target in neurodegenerative disease.
Collapse
Affiliation(s)
- Daniel Barnett
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY
| | - Till S. Zimmer
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Caroline Booraem
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY
| | - Fernando Palaguachi
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Samantha M. Meadows
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Edward T. Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Anna G. Orr
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY
| | - Adam L. Orr
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY
| |
Collapse
|
9
|
Liu R, Collier JM, Abdul-Rahman NH, Capuk O, Zhang Z, Begum G. Dysregulation of Ion Channels and Transporters and Blood-Brain Barrier Dysfunction in Alzheimer's Disease and Vascular Dementia. Aging Dis 2024; 15:1748-1770. [PMID: 38300642 PMCID: PMC11272208 DOI: 10.14336/ad.2023.1201] [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: 09/13/2023] [Accepted: 12/01/2023] [Indexed: 02/02/2024] Open
Abstract
The blood-brain barrier (BBB) plays a critical role in maintaining ion and fluid homeostasis, essential for brain metabolism and neuronal function. Regulation of nutrient, water, and ion transport across the BBB is tightly controlled by specialized ion transporters and channels located within its unique cellular components. These dynamic transport processes not only influence the BBB's structure but also impact vital signaling mechanisms, essential for its optimal function. Disruption in ion, pH, and fluid balance at the BBB is associated with brain pathology and has been implicated in various neurological conditions, including stroke, epilepsy, trauma, and neurodegenerative diseases such as Alzheimer's disease (AD). However, knowledge gaps exist regarding the impact of ion transport dysregulation on BBB function in neurodegenerative dementias. Several factors contribute to this gap: the complex nature of these conditions, historical research focus on neuronal mechanisms and technical challenges in studying the ion transport mechanisms in in vivo models and the lack of efficient in vitro BBB dementia models. This review provides an overview of current research on the roles of ion transporters and channels at the BBB and poses specific research questions: 1) How are the expression and activity of key ion transporters altered in AD and vascular dementia (VaD); 2) Do these changes contribute to BBB dysfunction and disease progression; and 3) Can restoring ion transport function mitigate BBB dysfunction and improve clinical outcomes. Addressing these gaps will provide a greater insight into the vascular pathology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ruijia Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jenelle M Collier
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | - Okan Capuk
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Zhongling Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Gulnaz Begum
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
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
|
11
|
Mahmood N, Choi JH, Wu PY, Dooling SW, Watkins TA, Huang Z, Lipman J, Zhao H, Yang A, Silversmith J, Inglebert Y, Koumenis C, Sharma V, Lacaille JC, Sossin WS, Khoutorsky A, McKinney RA, Costa-Mattioli M, Sonenberg N. The ISR downstream target ATF4 represses long-term memory in a cell type-specific manner. Proc Natl Acad Sci U S A 2024; 121:e2407472121. [PMID: 39047038 PMCID: PMC11295034 DOI: 10.1073/pnas.2407472121] [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: 04/14/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The integrated stress response (ISR), a pivotal protein homeostasis network, plays a critical role in the formation of long-term memory (LTM). The precise mechanism by which the ISR controls LTM is not well understood. Here, we report insights into how the ISR modulates the mnemonic process by using targeted deletion of the activating transcription factor 4 (ATF4), a key downstream effector of the ISR, in various neuronal and non-neuronal cell types. We found that the removal of ATF4 from forebrain excitatory neurons (but not from inhibitory neurons, cholinergic neurons, or astrocytes) enhances LTM formation. Furthermore, the deletion of ATF4 in excitatory neurons lowers the threshold for the induction of long-term potentiation, a cellular model for LTM. Transcriptomic and proteomic analyses revealed that ATF4 deletion in excitatory neurons leads to upregulation of components of oxidative phosphorylation pathways, which are critical for ATP production. Thus, we conclude that ATF4 functions as a memory repressor selectively within excitatory neurons.
Collapse
Affiliation(s)
- Niaz Mahmood
- Department of Biochemistry, McGill University, Montréal, QCH3A 1A3, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QCH3A 1A3, Canada
| | - Jung-Hyun Choi
- Department of Biochemistry, McGill University, Montréal, QCH3A 1A3, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QCH3A 1A3, Canada
| | - Pei You Wu
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QCH3G 0B1, Canada
| | - Sean W. Dooling
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
| | - Trent A. Watkins
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
| | - Ziying Huang
- Department of Biochemistry, McGill University, Montréal, QCH3A 1A3, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QCH3A 1A3, Canada
| | - Jesse Lipman
- Department of Biochemistry, McGill University, Montréal, QCH3A 1A3, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QCH3A 1A3, Canada
| | - Hanjie Zhao
- Department of Biochemistry, McGill University, Montréal, QCH3A 1A3, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QCH3A 1A3, Canada
| | - Anqi Yang
- Department of Biochemistry, McGill University, Montréal, QCH3A 1A3, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QCH3A 1A3, Canada
| | - Jake Silversmith
- Department of Biochemistry, McGill University, Montréal, QCH3A 1A3, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QCH3A 1A3, Canada
| | - Yanis Inglebert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QCH3G 0B1, Canada
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning, Research Group on Neural Signaling and Circuitry, University of Montréal, Montréal, QCH3T1J4, Canada
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104-5156
| | - Vijendra Sharma
- Department of Biomedical Sciences, University of Windsor, Windsor, ONN9B 3P4, Canada
| | - Jean-Claude Lacaille
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning, Research Group on Neural Signaling and Circuitry, University of Montréal, Montréal, QCH3T1J4, Canada
| | - Wayne S. Sossin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QCH3A 2B4, Canada
| | - Arkady Khoutorsky
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QCH4A3J1, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QCH3A 2B4, Canada
| | - R. Anne McKinney
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QCH3G 0B1, Canada
| | - Mauro Costa-Mattioli
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX77030
- Altos Labs Inc., Bay Area Institute of Science, Redwood City, CA94065
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montréal, QCH3A 1A3, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QCH3A 1A3, Canada
| |
Collapse
|
12
|
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
|
13
|
Bielanin JP, Metwally SAH, Oft HCM, Paruchuri SS, Lin L, Capuk O, Pennock ND, Song S, Sun D. NHE1 Protein in Repetitive Mild TBI-Mediated Neuroinflammation and Neurological Function Impairment. Antioxidants (Basel) 2024; 13:836. [PMID: 39061904 PMCID: PMC11274226 DOI: 10.3390/antiox13070836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Mild traumatic brain injuries (mTBIs) are highly prevalent and can lead to chronic behavioral and cognitive deficits often associated with the development of neurodegenerative diseases. Oxidative stress and formation of reactive oxygen species (ROS) have been implicated in mTBI-mediated axonal injury and pathogenesis. However, the underlying mechanisms and contributing factors are not completely understood. In this study, we explore these pathogenic mechanisms utilizing a murine model of repetitive mTBI (r-mTBI) involving five closed-skull concussions in young adult C57BL/6J mice. We observed a significant elevation of Na+/H+ exchanger protein (NHE1) expression in GFAP+ reactive astrocytes, IBA1+ microglia, and OLIG2+ oligodendrocytes across various brain regions (including the cerebral cortex, corpus callosum, and hippocampus) after r-mTBI. This elevation was accompanied by astrogliosis, microgliosis, and the accumulation of amyloid precursor protein (APP). Mice subjected to r-mTBI displayed impaired motor learning and spatial memory. However, post-r-mTBI administration of a potent NHE1 inhibitor, HOE642, attenuated locomotor and cognitive functional deficits as well as pathological signatures of gliosis, oxidative stress, axonal damage, and white matter damage. These findings indicate NHE1 upregulation plays a role in r-mTBI-induced oxidative stress, axonal damage, and gliosis, suggesting NHE1 may be a promising therapeutic target to alleviate mTBI-induced injuries and restore neurological function.
Collapse
Affiliation(s)
- John P. Bielanin
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Shamseldin A. H. Metwally
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Helena C. M. Oft
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Satya S. Paruchuri
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lin Lin
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Okan Capuk
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Nicholas D. Pennock
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Shanshan Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15213, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.P.B.); (S.A.H.M.); (H.C.M.O.); (S.S.P.); (L.L.); (O.C.); (N.D.P.); (S.S.)
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15213, USA
| |
Collapse
|
14
|
Diniz F, Parmeggiani B, Brandão G, Ferreira BK, Teixeira MF, Streck EL, Olivera-Bravo S, Barbeito LH, Schuck PF, de Melo Reis RA, Ferreira GC. Dual Effect of Carnosine on ROS Formation in Rat Cultured Cortical Astrocytes. Mol Neurobiol 2024; 61:4908-4922. [PMID: 38151612 DOI: 10.1007/s12035-023-03880-0] [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: 03/31/2023] [Accepted: 11/16/2023] [Indexed: 12/29/2023]
Abstract
Carnosine is composed of β-alanine and L-histidine and is considered to be an important neuroprotective agent with antioxidant, metal chelating, and antisenescence properties. However, children with serum carnosinase deficiency present increased circulating carnosine and severe neurological symptoms. We here investigated the in vitro effects of carnosine on redox and mitochondrial parameters in cultured cortical astrocytes from neonatal rats. Carnosine did not alter mitochondrial content or mitochondrial membrane potential. On the other hand, carnosine increased mitochondrial superoxide anion formation, levels of thiobarbituric acid reactive substances and oxidation of 2',7'-dichlorofluorescin diacetate (DCF-DA), indicating that carnosine per se acts as a pro-oxidant agent. Nonetheless, carnosine prevented DCF-DA oxidation induced by H2O2 in cultured cortical astrocytes. Since alterations on mitochondrial membrane potential are not likely to be involved in these effects of carnosine, the involvement of N-Methyl-D-aspartate (NMDA) receptors in the pro-oxidant actions of carnosine was investigated. MK-801, an antagonist of NMDA receptors, prevented DCF-DA oxidation induced by carnosine in cultured cortical astrocytes. Astrocyte reactivity induced by carnosine was also prevented by the coincubation with MK-801. The present study shows for the very first time the pro-oxidant effects of carnosine per se in astrocytes. The data raise awareness on the importance of a better understanding of the biological actions of carnosine, a nutraceutical otherwise widely reported as devoid of side effects.
Collapse
Affiliation(s)
- Fabiola Diniz
- Laboratório de Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Ciências Biológicas:Biofísica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, United States
| | - Belisa Parmeggiani
- Laboratório de Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriela Brandão
- Laboratório de Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna Klippel Ferreira
- Laboratório de Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Monique Fonseca Teixeira
- Laboratório de Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emilio Luiz Streck
- Laboratório de Doenças Neurometabólicas, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | | | | | - Patricia Fernanda Schuck
- Laboratório de Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Augusto de Melo Reis
- Programa de Pós-Graduação em Ciências Biológicas:Biofísica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Costa Ferreira
- Laboratório de Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas:Biofísica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Gong H, Xia Y, Jing G, Yuan M, Zhou H, Wu D, Zuo J, Lei C, Aidebaike D, Wu X, Song X. Berberine alleviates neuroinflammation by downregulating NFκB/LCN2 pathway in sepsis-associated encephalopathy: network pharmacology, bioinformatics, and experimental validation. Int Immunopharmacol 2024; 133:112036. [PMID: 38640713 DOI: 10.1016/j.intimp.2024.112036] [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: 01/25/2024] [Revised: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Sepsis refers to a systemic inflammatory response caused by infection, involving multiple organs. Sepsis-associated encephalopathy (SAE), as one of the most common complications in patients with severe sepsis, refers to the diffuse brain dysfunction caused by sepsis without central nervous system infection. However, there is no clear diagnostic criteria and lack of specific diagnostic markers. METHODS The main active ingredients of coptidis rhizoma(CR) were identified from TCMSP and SwissADME databases. SwissTargetPrediction and PharmMapper databases were used to obtain targets of CR. OMIM, DisGeNET and Genecards databases were used to explore targets of SAE. Limma differential analysis was used to identify the differential expressed genes(DEGs) in GSE167610 and GSE198861 datasets. WGCNA was used to identify feature module. GO and KEGG enrichment analysis were performed using Metascape, DAVID and STRING databases. The PPI network was constructed by STRING database and analyzed by Cytoscape software. AutoDock and PyMOL software were used for molecular docking and visualization. Cecal ligation and puncture(CLP) was used to construct a mouse model of SAE, and the core targets were verified in vivo experiments. RESULTS 277 common targets were identified by taking the intersection of 4730 targets related to SAE and 509 targets of 9 main active ingredients of CR. 52 common DEGs were mined from GSE167610 and GSE198861 datasets. Among the 25,864 DEGs in GSE198861, LCN2 showed the most significant difference (logFC = 6.9). GO and KEGG enrichment analysis showed that these 52 DEGs were closely related to "inflammatory response" and "innate immunity". A network containing 38 genes was obtained by PPI analysis, among which LCN2 ranked the first in Degree value. Molecular docking results showed that berberine had a well binding affinity with LCN2. Animal experiments results showed that berberine could inhibit the high expression of LCN2,S100A9 and TGM2 induced by CLP in the hippocampus of mice, as well as the high expression of inflammatory factors (TNFα, IL-6 and IL-1β). In addition, berberine might reduce inflammation and neuronal cell death by partially inhibiting NFκB/LCN2 pathway in the hippocampus of CLP models, thereby alleviating SAE. CONCLUSION Overall, Berberine may exert anti-inflammatory effects through multi-ingredients, multi-targets and multi-pathways to partially rescue neuronal death and alleviate SAE.
Collapse
Affiliation(s)
- Hailong Gong
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Yun Xia
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Guoqing Jing
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Huimin Zhou
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Die Wu
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Jing Zuo
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Chuntian Lei
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Delida Aidebaike
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China.
| | - Xuemin Song
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China.
| |
Collapse
|
16
|
Wang X, Li A, Fan H, Li Y, Yang N, Tang Y. Astrocyte-Derived Extracellular Vesicles for Ischemic Stroke: Therapeutic Potential and Prospective. Aging Dis 2024; 15:1227-1254. [PMID: 37728588 PMCID: PMC11081164 DOI: 10.14336/ad.2023.0823-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
Stroke is a leading cause of death and disability in the world. Astrocytes are special glial cells within the central nervous system and play important roles in mediating neuroprotection and repair processes during stroke. Extracellular vesicles (EVs) are lipid bilayer particles released from cells that facilitate intercellular communication in stroke by delivering proteins, lipids, and RNA to target cells. Recently, accumulating evidence suggested that astrocyte-derived EVs (ADEVs) are actively involved in mediating numerous biological processes including neuroprotection and neurorepair in stroke and they are realized as an excellent therapeutic approach for treating stroke. In this review we systematically summarize the up-to-date research on ADEVs in stroke, and prospects for its potential as a novel therapeutic target for stroke. We also provide an overview of the effects and functions of ADEVs on stroke recovery, which may lead to developing clinically relevant therapies for stroke.
Collapse
Affiliation(s)
- Xianghui Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China.
- School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Aihua Li
- Department of rehabilitation medicine, Jinan Hospital, Jinan, China
| | - Huaju Fan
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China.
| | - Yanyan Li
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China.
| | - Nana Yang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China.
- School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Yaohui Tang
- School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
17
|
Cao H, Li B, Mu M, Li S, Chen H, Tao H, Wang W, Zou Y, Zhao Y, Liu Y, Tao X. Nicotine suppresses crystalline silica-induced astrocyte activation and neuronal death by inhibiting NF-κB in the mouse hippocampus. CNS Neurosci Ther 2024; 30:e14508. [PMID: 37864452 PMCID: PMC11017465 DOI: 10.1111/cns.14508] [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: 06/22/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/22/2023] Open
Abstract
AIMS Exposure to crystalline silica (CS) in occupational settings induces chronic inflammation in the respiratory system and, potentially, the brain. Some workers are frequently concurrently exposed to both CS and nicotine. Here, we explored the impact of nicotine on CS-induced neuroinflammation in the mouse hippocampus. METHODS In this study, we established double-exposed models of CS and nicotine in C57BL/6 mice. To assess depression-like behavior, experiments were conducted at 3, 6, and 9 weeks. Serum inflammatory factors were analyzed by ELISA. Hippocampus was collected for RNA sequencing analysis and examining the gene expression patterns linked to inflammation and cell death. Microglia and astrocyte activation and hippocampal neuronal death were assessed using immunohistochemistry and immunofluorescence staining. Western blotting was used to analyze the NF-κB expression level. RESULTS Mice exposed to CS for 3 weeks showed signs of depression. This was accompanied by elevated IL-6 in blood, destruction of the blood-brain barrier, and activation of astrocytes caused by an increased NF-κB expression in the CA1 area of the hippocampus. The elevated levels of astrocyte-derived Lcn2 and upregulated genes related to inflammation led to higher neuronal mortality. Moreover, nicotine mitigated the NF-κB expression, astrocyte activation, and neuronal death, thereby ameliorating the associated symptoms. CONCLUSION Silica exposure induces neuroinflammation and neuronal death in the mouse hippocampal CA1 region and depressive behavior. However, nicotine inhibits CS-induced neuroinflammation and neuronal apoptosis, alleviating depressive-like behaviors in mice.
Collapse
Affiliation(s)
- Hangbing Cao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Bing Li
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Min Mu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Shanshan Li
- School of PharmacyBengbu Medical CollegeBengbuChina
| | - Haoming Chen
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Huihui Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Wenyang Wang
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yuanjie Zou
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yehong Zhao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Yang Liu
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| | - Xinrong Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of EducationAnhui University of Science and TechnologyHuainanChina
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education InstitutesAnhui University of Science and TechnologyHuainanChina
- Anhui Province Engineering Laboratory of Occupational Health and SafetyAnhui University of Science and TechnologyHuainanChina
- School of Medicine, Department of Medical Frontier Experimental CenterAnhui University of Science and TechnologyHuainanChina
| |
Collapse
|
18
|
Tan Q, Zhang C, Rao X, Wan W, Lin W, Huang S, Ying J, Lin Y, Hua F. The interaction of lipocalin-2 and astrocytes in neuroinflammation: mechanisms and therapeutic application. Front Immunol 2024; 15:1358719. [PMID: 38533497 PMCID: PMC10963420 DOI: 10.3389/fimmu.2024.1358719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
Neuroinflammation is a common pathological process in various neurological disorders, including stroke, Alzheimer's disease, Parkinson's disease, and others. It involves the activation of glial cells, particularly astrocytes, and the release of inflammatory mediators. Lipocalin-2 (Lcn-2) is a secretory protein mainly secreted by activated astrocytes, which can affect neuroinflammation through various pathways. It can also act as a pro-inflammatory factor by modulating astrocyte activation and polarization through different signaling pathways, such as NF-κB, and JAK-STAT, amplifying the inflammatory response and aggravating neural injury. Consequently, Lcn-2 and astrocytes may be potential therapeutic targets for neuroinflammation and related diseases. This review summarizes the current knowledge on the role mechanisms, interactions, and therapeutic implications of Lcn-2 and astrocytes in neuroinflammation.
Collapse
Affiliation(s)
- Qianqian Tan
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chenxi Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiuqin Rao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Wan
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Lin
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shupeng Huang
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Gu M, Li X, Wu R, Cheng X, Zhou S, Gu X. The Transcription Factor Ets1 Influences Axonal Growth via Regulation of Lcn2. Mol Neurobiol 2024; 61:971-981. [PMID: 37672148 PMCID: PMC10861751 DOI: 10.1007/s12035-023-03616-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 08/25/2023] [Indexed: 09/07/2023]
Abstract
Transcription factors are essential for the development and regeneration of the nervous system. The current study investigated key regulatory transcription factors in rat spinal cord development via RNA sequencing. The hub gene Ets1 was highly expressed in the spinal cord during the embryonic period, and then its expression decreased during spinal cord development. Knockdown of Ets1 significantly increased the axonal growth of cultured spinal cord neurons. Luciferase reporter assays and chromatin immunoprecipitation assays indicated that Ets1 could directly bind to the Lcn2 promoter and positively regulate Lcn2 transcription. In conclusion, these findings provide the first direct evidence that Ets1 regulates axon growth by controlling Lcn2 expression, and Ets1 may be a novel therapeutic target for axon regeneration in the central nervous system.
Collapse
Affiliation(s)
- Miao Gu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- School of Basic Medical Sciences, Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde, Hebei, China
| | - Xiaodi Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ronghua Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Xiao Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| | - Xiaosong Gu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
20
|
Beltran-Lobo P, Hughes MM, Troakes C, Croft CL, Rupawala H, Jutzi D, Ruepp MD, Jimenez-Sanchez M, Perkinton MS, Kassiou M, Golde TE, Hanger DP, Verkhratsky A, Perez-Nievas BG, Noble W. P2X 7R influences tau aggregate burden in human tauopathies and shows distinct signalling in microglia and astrocytes. Brain Behav Immun 2023; 114:414-429. [PMID: 37716378 PMCID: PMC10896738 DOI: 10.1016/j.bbi.2023.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 09/18/2023] Open
Abstract
The purinoceptor P2X7R is a promising therapeutic target for tauopathies, including Alzheimer's disease (AD). Pharmacological inhibition or genetic knockdown of P2X7R ameliorates cognitive deficits and reduces pathological tau burden in mice that model aspects of tauopathy, including mice expressing mutant human frontotemporal dementia (FTD)-causing forms of tau. However, disagreements remain over which glial cell types express P2X7R and therefore the mechanism of action is unresolved. Here, we show that P2X7R protein levels increase in human AD post-mortem brain, in agreement with an upregulation of P2RX7 mRNA observed in transcriptome profiles from the AMP-AD consortium. P2X7R protein increases mirror advancing Braak stage and coincide with synapse loss. Using RNAScope we detect P2RX7 mRNA in microglia and astrocytes in human AD brain, including in the vicinity of senile plaques. In cultured microglia, P2X7R activation modulates the NLRP3 inflammasome pathway by promoting the formation of active complexes and release of IL-1β. In astrocytes, P2X7R activates NFκB signalling and increases production of the cytokines CCL2, CXCL1 and IL-6 together with the acute phase protein Lcn2. To further explore the role of P2X7R in a disease-relevant context, we expressed wild-type or FTD-causing mutant forms of tau in mouse organotypic brain slice cultures. Inhibition of P2X7R reduces insoluble tau levels without altering soluble tau phosphorylation or synaptic localisation, suggesting a non-cell autonomous role of glial P2X7R on pathological tau aggregation. These findings support further investigations into the cell-type specific effects of P2X7R-targeting therapies in tauopathies.
Collapse
Affiliation(s)
- Paula Beltran-Lobo
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK
| | - Martina M Hughes
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK
| | - Claire Troakes
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK; London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Cara L Croft
- UK Dementia Research Institute, UCL Institute of Neurology, University College London, London, UK; The Francis Crick Institute, London, UK
| | - Huzefa Rupawala
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK
| | - Daniel Jutzi
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK; UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Marc-David Ruepp
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK; UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Maria Jimenez-Sanchez
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK
| | | | - Michael Kassiou
- School of Chemistry, Faculty of Science, University of Sydney, Sydney, New South Wales, Australia
| | - Todd E Golde
- Department of Pharmacology and Chemical Biology, Department of Neurology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
| | - Diane P Hanger
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Beatriz G Perez-Nievas
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK.
| | - Wendy Noble
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London SE5 9RX, UK; University of Exeter, Department of Clinical and Biomedical Science, Hatherly Laboratories, Prince of Wales Road, Exeter EX4 4PS, UK.
| |
Collapse
|
21
|
Zhou M, Xin J, Chen J, Sun C, Huo B, Zhang W, Liu X. Scientific Landscape of Oxidative Stress in Stroke: From a Bibliometric Analysis to an in-Depth Review. Neurochem Res 2023; 48:3327-3348. [PMID: 37505366 DOI: 10.1007/s11064-023-03999-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Stroke is an acute cerebrovascular disease resulting from either obstruction or rupture of a blood vessel in the brain. Oxidative stress (OS), referred to a status where cellular oxidative capacities overwhelm antioxidative defenses, is involved in the pathophysiology of stroke. The bibliometric analysis and in-depth review aim to depict the research trend of OS in stroke. Relevant scientific publications were acquired from the Web of Science Core Collection database. Scientific landscape of OS in stroke was illustrated by general quantitative trend, impactful journals, and co-authorship of various academic units (i.e., countries/regions, organizations, and authors). Furthermore, theme analysis predicting the hot research issues and frontiers was performed. 15,826 documents regarding OS in stroke were obtained over a time span of more than 20 years from 1992 to 2021. The overall tendency of publication counts was continuously on the rise. Bibliometric analysis indicated China and the United States were predominant in this study field, as reflected by their high publication counts and intensive collaboration with other countries. Current key research areas of OS in stroke may lie in the investigation of neuroinflammation, and interaction among multiple cell death mechanisms including apoptosis, autophagy, and ferroptosis to search for effective treatments. Moreover, another hot topic could be the association between air pollution and stroke, and its underlying mechanisms. As the exploration of OS in stroke is speculated to be a continuous hot spot in the future, this article may be helpful for researchers to conduct future studies with the understanding of influential academic forces and research highlights.
Collapse
Affiliation(s)
- Minqi Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China
| | - Jiayu Xin
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China
| | - Jinyu Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Caiyun Sun
- Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Bingyue Huo
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China
| | - Wenting Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China
| | - Xiangqian Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hang Kong Road, Wuhan, 430030, China.
| |
Collapse
|
22
|
Jiang SY, Tian T, Yao H, Xia XM, Wang C, Cao L, Hu G, Du RH, Lu M. The cGAS-STING-YY1 axis accelerates progression of neurodegeneration in a mouse model of Parkinson's disease via LCN2-dependent astrocyte senescence. Cell Death Differ 2023; 30:2280-2292. [PMID: 37633968 PMCID: PMC10589362 DOI: 10.1038/s41418-023-01216-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023] Open
Abstract
Recent studies provide clues that astrocyte senescence is correlated with Parkinson's disease (PD) progression, while little is known about the molecular basis for astrocyte senescence in PD. Here, we found that cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) was upregulated in senescent astrocytes of PD and aged mice. Strikingly, deletion of astrocytic cGAS significantly prevented senescence of astrocytes and neurodegeneration. Furthermore, we identified LCN2 as the effector of cGAS-STING signal by RNA-Seq analysis. Genetic manipulation of LCN2 expression proved the regulation of cGAS-STING-LCN2 axis in astrocyte senescence. Additionally, YY1 was discovered as the transcription factor of LCN2 by chromatin immunoprecipitation. Binding of STING to YY1 impedes nuclear translocation of YY1. Herein, we determine the involvement of the cGAS-STING-YY1-LCN2 signaling cascade in the control of astrocyte senescence and PD progression. Together, this work fills the gap in our understanding of astrocyte senescence, and provides potential targets for delaying PD progression.
Collapse
Affiliation(s)
- Si-Yuan Jiang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China
| | - Tian Tian
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China
| | - Hang Yao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China
| | - Xiao-Mei Xia
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China
| | - Cong Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China
| | - Lei Cao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China
| | - Ren-Hong Du
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, PR China.
| |
Collapse
|
23
|
Long H, Zhu W, Wei L, Zhao J. Iron homeostasis imbalance and ferroptosis in brain diseases. MedComm (Beijing) 2023; 4:e298. [PMID: 37377861 PMCID: PMC10292684 DOI: 10.1002/mco2.298] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 06/29/2023] Open
Abstract
Brain iron homeostasis is maintained through the normal function of blood-brain barrier and iron regulation at the systemic and cellular levels, which is fundamental to normal brain function. Excess iron can catalyze the generation of free radicals through Fenton reactions due to its dual redox state, thus causing oxidative stress. Numerous evidence has indicated brain diseases, especially stroke and neurodegenerative diseases, are closely related to the mechanism of iron homeostasis imbalance in the brain. For one thing, brain diseases promote brain iron accumulation. For another, iron accumulation amplifies damage to the nervous system and exacerbates patients' outcomes. In addition, iron accumulation triggers ferroptosis, a newly discovered iron-dependent type of programmed cell death, which is closely related to neurodegeneration and has received wide attention in recent years. In this context, we outline the mechanism of a normal brain iron metabolism and focus on the current mechanism of the iron homeostasis imbalance in stroke, Alzheimer's disease, and Parkinson's disease. Meanwhile, we also discuss the mechanism of ferroptosis and simultaneously enumerate the newly discovered drugs for iron chelators and ferroptosis inhibitors.
Collapse
Affiliation(s)
- Haining Long
- Department of Diagnostic and Interventional RadiologyShanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School
of MedicineShanghaiChina
| | - Wangshu Zhu
- Department of Diagnostic and Interventional RadiologyShanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School
of MedicineShanghaiChina
| | - Liming Wei
- Department of Diagnostic and Interventional RadiologyShanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School
of MedicineShanghaiChina
| | - Jungong Zhao
- Department of Diagnostic and Interventional RadiologyShanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School
of MedicineShanghaiChina
| |
Collapse
|
24
|
Guo L, Hu C, Yao M, Han G. Mechanism of sorafenib resistance associated with ferroptosis in HCC. Front Pharmacol 2023; 14:1207496. [PMID: 37351514 PMCID: PMC10282186 DOI: 10.3389/fphar.2023.1207496] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/26/2023] [Indexed: 06/24/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most familiar primary hepatic malignancy with a poor prognosis. The incidence of HCC and the associated deaths have risen in recent decades. Sorafenib is the first drug to be approved by the Food and Drug Administration (FDA) for routine use in the first-line therapy of patients with advanced HCC. However, only about 30% of patients with HCC will be benefited from sorafenib therapy, and drug resistance typically develops within 6 months. In recent years, the mechanisms of resistance to sorafenib have gained the attention of a growing number of researchers. A promising field of current studies is ferroptosis, which is a novel form of cell death differing from apoptosis, necroptosis, and autophagy. This process is dependent on the accumulation of intracellular iron and reactive oxygen species (ROS). Furthermore, the increase in intracellular iron levels and ROS can be significantly observed in cells resistant to sorafenib. This article reviews the mechanisms of resistance to sorafenib that are related to ferroptosis, evaluates the relationship between ferroptosis and sorafenib resistance, and explores new therapeutic approaches capable of reversing sorafenib resistance in HCC through the modulation of ferroptosis.
Collapse
|
25
|
Furutani K, Chen O, McGinnis A, Wang Y, Serhan CN, Hansen TV, Ji RR. Novel proresolving lipid mediator mimetic 3-oxa-PD1n-3 docosapentaenoic acid reduces acute and chronic itch by modulating excitatory and inhibitory synaptic transmission and astroglial secretion of lipocalin-2 in mice. Pain 2023; 164:1340-1354. [PMID: 36378290 PMCID: PMC10182233 DOI: 10.1097/j.pain.0000000000002824] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
ABSTRACT Specialized proresolving mediators (SPMs) have demonstrated potent analgesic actions in animal models of pathological pain. The actions of SPMs in acute and chronic itch are currently unknown. Recently, n-3 docosapentaenoic acid (DPA) was found to be a substrate for the biosynthesis of several novel families of SPMs and 3-oxa-PD1 n-3 DPA (3-oxa-PD1) is an oxidation-resistant metabolic stable analogue of the n-3 DPA-derived protectin D1 (PD1). In this article, we demonstrate that 3-oxa-PD1 effectively reduces both acute and chronic itch in mouse models. Intrathecal injection of 3-oxa-PD1 (100 ng) reduced acute itch induced by histamine, chloroquine, or morphine. Furthermore, intrathecal 3-oxa-PD1 effectively reduced chronic itch, induced by cutaneous T-cell lymphoma (CTCL), allergic contact dermatitis with dinitrofluorobenzene, and psoriasis by imiquimod. Intratumoral injection of 3-oxa-PD1 also suppressed CTCL-induced chronic itch. Strikingly, the antipruritic effect lasted for several weeks after 1-week intrathecal 3-oxa-PD1 treatment. Whole-cell recordings revealed significant increase in excitatory postsynaptic currents in spinal dorsal horn (SDH) neurons of CTCL mice, but this increase was blocked by 3-oxa-PD1. 3-oxa-PD1 further increased inhibitory postsynaptic currents in SDH neurons of CTCL mice. Cutaneous T-cell lymphoma increased the spinal levels of lipocalin-2 (LCN2), an itch mediator produced by astrocytes. 3-oxa-PD1 suppressed LCN2 production in CTCL mice and LCN2 secretion in astrocytes. Finally, CTCL-induced anxiety was alleviated by intrathecal 3-oxa-PD1. Our findings suggest that 3-oxa-PD1 potently inhibits acute and chronic itch through the regulation of excitatory or inhibitory synaptic transmission and astroglial LCN2 production. Therefore, stable SPM analogs such as 3-oxa-PD1 could be useful to treat pruritus associated with different skin injuries.
Collapse
Affiliation(s)
- Kenta Furutani
- Center for Translational Pain Medicine, Department of Anesthesiology, and Department of Neurobiology, Duke University Medical Center, Durham, NC 27710
| | - Ouyang Chen
- Center for Translational Pain Medicine, Department of Anesthesiology, and Department of Neurobiology, Duke University Medical Center, Durham, NC 27710
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710
| | - Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, and Department of Neurobiology, Duke University Medical Center, Durham, NC 27710
| | - Yuqing Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, and Department of Neurobiology, Duke University Medical Center, Durham, NC 27710
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Hale Building for Transformative Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, 02115
| | - Trond Vidar Hansen
- Department of Pharmacy, Section for Pharmaceutical Chemistry, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, and Department of Neurobiology, Duke University Medical Center, Durham, NC 27710
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
26
|
Kaur N, LaForce G, Mallela DP, Saha PP, Buffa J, Li XS, Sangwan N, Rothenberg K, Zhu W. Exploratory Transcriptomic Profiling Reveals the Role of Gut Microbiota in Vascular Dementia. Int J Mol Sci 2023; 24:ijms24098091. [PMID: 37175797 PMCID: PMC10178712 DOI: 10.3390/ijms24098091] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Stroke is the second most common cause of cognitive impairment and dementia. Vascular dementia (VaD), a cognitive impairment following a stroke, is common and significantly impacts the quality of life. We recently demonstrated via gut microbe transplant studies that the gut microbe-dependent trimethylamine-N-oxide (TMAO) pathway impacts stroke severity, both infarct size and long-term cognitive outcomes. However, the molecular mechanisms that underly the role of the microbiome in VaD have not been explored in depth. To address this issue, we performed a comprehensive RNA-sequencing analysis to identify differentially expressed (DE) genes in the ischemic cerebral cortex of mouse brains at pre-stroke and post-stroke day 1 and day 3. A total of 4016, 3752 and 7861 DE genes were identified at pre-stroke and post-stroke day 1 and day 3, respectively. The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated pathways of neurodegeneration in multiple diseases, chemokine signaling, calcium signaling, and IL-17 signaling as the key enriched pathways. Inflammatory response genes interleukin-1 beta (Il-1β), chemokines (C-X-C motif chemokine ligand 10 (Cxcl10), chemokine ligand 2 (Ccl2)), and immune system genes (S100 calcium binding protein 8 (S100a8), lipocalin-2 (Lcn2)) were among the most significantly upregulated genes. Hypocretin neuropeptide precursor (Hcrt), a neuropeptide, and transcription factors such as neuronal PAS domain protein 4 (Npas4), GATA binding protein 3 (Gata3), and paired box 7 (Pax7) were among the most significantly downregulated genes. In conclusion, our results indicate that higher plasma TMAO levels induce differential mRNA expression profiles in the ischemic brain tissue in our pre-clinical stroke model, and the predicted pathways provide the molecular basis for regulating the TMAO-enhanced neuroinflammatory response in the brain.
Collapse
Affiliation(s)
- Navdeep Kaur
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Geneva LaForce
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Deepthi P Mallela
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Prasenjit Prasad Saha
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jennifer Buffa
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Naseer Sangwan
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
- Microbial Sequencing & Analytics Resource (MSAAR) Facility, Shared Laboratory Resources (SLR), Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kasia Rothenberg
- Cleveland Clinic Lou Ruvo Center for Brain Health, Cleveland, OH 44195, USA
| | - Weifei Zhu
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
27
|
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: 12] [Impact Index Per Article: 12.0] [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
|
28
|
Rossi G, Ordazzo G, Vanni NN, Castoldi V, Iannielli A, Di Silvestre D, Bellini E, Bernardo L, Giannelli SG, Luoni M, Muggeo S, Leocani L, Mauri P, Broccoli V. MCT1-dependent energetic failure and neuroinflammation underlie optic nerve degeneration in Wolfram syndrome mice. eLife 2023; 12:81779. [PMID: 36645345 PMCID: PMC9891717 DOI: 10.7554/elife.81779] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Wolfram syndrome 1 (WS1) is a rare genetic disorder caused by mutations in the WFS1 gene leading to a wide spectrum of clinical dysfunctions, among which blindness, diabetes, and neurological deficits are the most prominent. WFS1 encodes for the endoplasmic reticulum (ER) resident transmembrane protein wolframin with multiple functions in ER processes. However, the WFS1-dependent etiopathology in retinal cells is unknown. Herein, we showed that Wfs1 mutant mice developed early retinal electrophysiological impairments followed by marked visual loss. Interestingly, axons and myelin disruption in the optic nerve preceded the degeneration of the retinal ganglion cell bodies in the retina. Transcriptomics at pre-degenerative stage revealed the STAT3-dependent activation of proinflammatory glial markers with reduction of the homeostatic and pro-survival factors glutamine synthetase and BDNF. Furthermore, label-free comparative proteomics identified a significant reduction of the monocarboxylate transport isoform 1 (MCT1) and its partner basigin that are highly enriched on retinal glia and myelin-forming oligodendrocytes in optic nerve together with wolframin. Loss of MCT1 caused a failure in lactate transfer from glial to neuronal cell bodies and axons leading to a chronic hypometabolic state. Thus, this bioenergetic impairment is occurring concurrently both within the axonal regions and cell bodies of the retinal ganglion cells, selectively endangering their survival while impacting less on other retinal cells. This metabolic dysfunction occurs months before the frank RGC degeneration suggesting an extended time-window for intervening with new therapeutic strategies focused on boosting retinal and optic nerve bioenergetics in WS1.
Collapse
Affiliation(s)
- Greta Rossi
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Gabriele Ordazzo
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Niccolò N Vanni
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Valerio Castoldi
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), San Raffaele Scientific InstituteMilanItaly
| | - Angelo Iannielli
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- National Research Council of Italy, Institute of NeuroscienceMilanoItaly
| | - Dario Di Silvestre
- National Research Council of Italy, Institute of Technologies in BiomedicineMilanItaly
| | - Edoardo Bellini
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Letizia Bernardo
- National Research Council of Italy, Institute of Technologies in BiomedicineMilanItaly
| | | | - Mirko Luoni
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- National Research Council of Italy, Institute of NeuroscienceMilanoItaly
| | - Sharon Muggeo
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Letizia Leocani
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), San Raffaele Scientific InstituteMilanItaly
| | - PierLuigi Mauri
- National Research Council of Italy, Institute of Technologies in BiomedicineMilanItaly
| | - Vania Broccoli
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- National Research Council of Italy, Institute of NeuroscienceMilanoItaly
| |
Collapse
|