51
|
Jia YJ, Li QS. Ferroptosis: a critical player and potential therapeutic target in traumatic brain injury and spinal cord injury. Neural Regen Res 2023; 18:506-512. [DOI: 10.4103/1673-5374.350187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
52
|
D'Mello V, Subramaniam M, Bhalla AP, Saavedra S, Leiba O, Levison SW. Intranasal Leukemia Inhibitory Factor Attenuates Gliosis and Axonal Injury and Improves Sensorimotor Function After a Mild Pediatric Traumatic Brain Injury. Neurotrauma Rep 2023; 4:236-250. [PMID: 37095853 PMCID: PMC10122240 DOI: 10.1089/neur.2021.0075] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Leukemia inhibitory factor (LIF) is a neuroprotective cytokine that is essential for appropriate glial responses, remyelination, and preservation of neuronal conductance after injury. The intranasal route for delivery of therapeutics to the central nervous system is of particular interest given that it bypasses the blood-brain barrier and peripheral clearance systems. We explored the possibility that LIF might improve neurological function when administered intranasally during the acute phase in a pediatric model of mild traumatic brain injury (mTBI). We tested two doses of LIF and evaluated behavioral outcomes. Here, we show that acute 40-ng intranasal LIF treatment twice a day for 3 days attenuates astrogliosis and microgliosis, protects against axonal damage, significantly improves sensorimotor function, and is well tolerated without detrimental effects on growth. Altogether, our studies provide pre-clinical evidence for the use of acute intranasal LIF treatment as a viable therapeutic for pediatric cases of mTBIs.
Collapse
Affiliation(s)
- Veera D'Mello
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Malini Subramaniam
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Aditya Paul Bhalla
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Sherlyn Saavedra
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Ofri Leiba
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Steven W. Levison
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- Address correspondence to: Steven W. Levison, PhD, Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, 205 South Orange Avenue, Newark, NJ 07103, USA.
| |
Collapse
|
53
|
Schirinzi T, Maftei D, Passali FM, Grillo P, Zenuni H, Mascioli D, Maurizi R, Loccisano L, Vincenzi M, Rinaldi AM, Ralli M, Di Girolamo S, Stefani A, Lattanzi R, Severini C, Mercuri NB. Olfactory Neuron Prokineticin-2 as a Potential Target in Parkinson's Disease. Ann Neurol 2023; 93:196-204. [PMID: 36218142 DOI: 10.1002/ana.26526] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The objective of this study was to outline the dynamics of prokineticin-2 pathway in relation to clinical-pathological features of Parkinson's disease by examining olfactory neurons of patients. METHODS Thirty-eight patients (26 de novo, newly diagnosed) and 31 sex/age-matched healthy controls underwent noninvasive mucosa brushing for olfactory neurons collection, and standard clinical assessment. Gene expression levels of prokineticin-2, prokineticin-2 receptors type 1 and 2, and prokineticin-2-long peptide were measured in olfactory neurons by real-time polymerase chain reaction (PCR); moreover, the prokineticin-2 protein and α-synuclein species (total and oligomeric) were quantified by immunofluorescence staining. RESULTS Prokineticin-2 expression was significantly increased in Parkinson's disease. De novo patients had higher prokineticin-2 levels, directly correlated with Movement Disorder Society-Sponsored Revision of the Unified Parkinson Disease Rating Scale (MDS-UPDRS) part III motor score. In addition, oligomeric α-synuclein was higher in Parkinson's disease and directly correlated with prokineticin-2 protein levels. Total α-synuclein did not differ between patients and controls. INTERPRETATION Prokineticin-2 is a chemokine showing neuroprotective effects in experimental models of Parkinson's disease, but translational proof of its role in patients is still lacking. Here, we used olfactory neurons as the ideal tissue to analyze molecular stages of neurodegeneration in vivo, providing unprecedented evidence that the prokineticin-2 pathway is activated in patients with Parkinson's disease. Specifically, prokineticin-2 expression in olfactory neurons was higher at early disease stages, proportional to motor severity, and associated with oligomeric α-synuclein accumulation. These data, consistently with preclinical findings, support prokineticin-2 as a candidate target in Parkinson's disease, and validate reliability of olfactory neurons to reflect pathological changes of the disease. ANN NEUROL 2023;93:196-204.
Collapse
Affiliation(s)
- Tommaso Schirinzi
- Unit of Neurology, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Daniela Maftei
- Department of Physiology and Pharmacology "V. Erspamer,", Sapienza University of Rome, Rome, Italy
| | - Francesco M Passali
- Unit of ENT, Department of Clinical Sciences and Translational Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Piergiorgio Grillo
- Unit of Neurology, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Henri Zenuni
- Unit of Neurology, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Davide Mascioli
- Unit of Neurology, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Riccardo Maurizi
- Unit of ENT, Department of Clinical Sciences and Translational Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Laura Loccisano
- Unit of ENT, Department of Clinical Sciences and Translational Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Martina Vincenzi
- Department of Physiology and Pharmacology "V. Erspamer,", Sapienza University of Rome, Rome, Italy
| | - Anna Maria Rinaldi
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Stefano Di Girolamo
- Unit of ENT, Department of Clinical Sciences and Translational Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Alessandro Stefani
- Unit of Neurology, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Roberta Lattanzi
- Department of Physiology and Pharmacology "V. Erspamer,", Sapienza University of Rome, Rome, Italy
| | - Cinzia Severini
- Department of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Nicola B Mercuri
- Unit of Neurology, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy.,European Centre for Brain Research, IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
54
|
Wu Z, Sun J, Liao Z, Qiao J, Chen C, Ling C, Wang H. An update on the therapeutic implications of long-chain acyl-coenzyme A synthetases in nervous system diseases. Front Neurosci 2022; 16:1030512. [PMID: 36507355 PMCID: PMC9731139 DOI: 10.3389/fnins.2022.1030512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Long-chain acyl-coenzyme A synthetases (ACSLs) are a family of CoA synthetases that activate fatty acid (FA) with chain lengths of 12-20 carbon atoms by forming the acyl-AMP derivative in an isozyme-specific manner. This family mainly includes five members (ACSL1, ACSL3, ACSL4, ACSL5, and ACSL6), which are thought to have specific and different functions in FA metabolism and oxidative stress of mammals. Accumulating evidence shows that the dysfunction of ACSLs is likely to affect cell proliferation and lead to metabolic diseases in multiple organs and systems through different signaling pathways and molecular mechanisms. Hence, a central theme of this review is to emphasize the therapeutic implications of ACSLs in nervous system disorders.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi Liao
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia Qiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China,*Correspondence: Hui Wang,
| |
Collapse
|
55
|
Nie Z, Tan L, Niu J, Wang B. The role of regulatory necrosis in traumatic brain injury. Front Mol Neurosci 2022; 15:1005422. [PMID: 36329694 PMCID: PMC9622788 DOI: 10.3389/fnmol.2022.1005422] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability in the population worldwide, of which key injury mechanism involving the death of nerve cells. Many recent studies have shown that regulatory necrosis is involved in the pathological process of TBI which includes necroptosis, pyroptosis, ferroptosis, parthanatos, and Cyclophilin D (CypD) mediated necrosis. Therefore, targeting the signaling pathways involved in regulatory necrosis may be an effective strategy to reduce the secondary injury after TBI. Meanwhile, drugs or genes are used as interference factors in various types of regulatory necrosis, so as to explore the potential treatment methods for the secondary injury after TBI. This review summarizes the current progress on regulatory necrosis in TBI.
Collapse
|
56
|
Meng Y, Sun H, Li Y, Zhao S, Su J, Zeng F, Deng G, Chen X. Targeting Ferroptosis by Ubiquitin System Enzymes: A Potential Therapeutic Strategy in Cancer. Int J Biol Sci 2022; 18:5475-5488. [PMID: 36147464 PMCID: PMC9461661 DOI: 10.7150/ijbs.73790] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Ferroptosis is a novel type of regulated cell death driven by the excessive accumulation of iron-dependent lipid peroxidation. Therapy-resistant tumor cells, particularly those in the mesenchymal-like state and prone to metastasis, are highly susceptible to ferroptosis, suggesting that induction of ferroptosis in tumor cells is a promising strategy for cancer therapy. Although ferroptosis is regulated at various levels, ubiquitination is key to post-translational regulation of ferroptotic cell death. E3 ubiquitin ligases (E3s) and deubiquitinating enzymes (DUBs) are the most remarkable ubiquitin system enzymes, whose dysregulation accounts for the progression of multiple cancers. E3s are involved in the attachment of ubiquitin to substrates for their degradation, and this process is reversed by DUBs. Accumulating evidence has highlighted the important role of ubiquitin system enzymes in regulating the sensitivity of ferroptosis. Herein, we will portray the regulatory networks of ferroptosis mediated by E3s or DUBs and discuss opportunities and challenges for incorporating this regulation into cancer therapy.
Collapse
Affiliation(s)
- Yu Meng
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huiyan Sun
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yayun Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuang Zhao
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Su
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Furong Zeng
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangtong Deng
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
57
|
Prokineticins as a Prognostic Biomarker for Low-Grade Gliomas: A Study Based on The Cancer Genome Atlas Data. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2309339. [PMID: 35845958 PMCID: PMC9283042 DOI: 10.1155/2022/2309339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
Lower-grade glioma (LGG) is a crucial pathological type of glioma. Prokineticins have not been reported in LGG. Prokineticins as a member of the multifunctional chemokine-like peptide family are divided into two ligands: PROK1 and PROK2. We evaluated the role of PROK1 and PROK2 in LGG using TCGA database. We downloaded the datasets of LGG from TCGA and evaluated the influence of prokineticins on LGG survival by survival module. Correlations between clinical information and prokineticins expression were analyzed using logistic regression. Univariable survival and multivariate Cox analysis was used to compare several clinical characteristics with survival. Correlation between prokineticins and cancer immune infiltrates was explored using CIBERSORT and correlation module of GEPIA. We analyzed genes of PROK1 and PROK2 affecting LGG, screened differentially expressed genes (DEGs), interacted protein-protein with DEGs through the STRING website, then imported the results into the Cytospace software, and calculated the hub genes. To analyze whether hub genes and prokineticins are related, the relationship between PROK1 and PROK2 and hub genes was assessed and shown by heat map. In addition, gene set enrichment analysis (GSEA) was performed using the TCGA dataset. The univariate analysis using logistic regression and PROK1 and PROK2 showed opposite expression differences between tumor and normal tissues (
). PRO1 and PROK2 expressions showed significant differences in tumor grade, age, Iiscitrate DeHydrogenase (IDH) status, histological type, and 1P/19q codeletion. Multivariate analysis revealed that the up-regulated PROK1 and PROK2 expression is an independent prognostic factor for bad prognosis. Specifically, prokineticin expression level has significant correlations with infiltrating levels of Th1 cells, NK CD 56bright cells, and Mast cells in LGG. We screened 21 DEGs and obtained 5 hub genes (HOXC10, HOXD13, SOX4, GATA4, HOXA9). GSEA-identified FCMR activation, creation of C4 and C2 activators, and CD22-mediated BCR regulation in gene ontology (GO) were differentially enriched in high PROK1 and PROK2 expression phenotype pathway, cytoplasmic ribosomal proteins, and ribosome and were differentially enriched in the low PROK1 and PROK2 expression phenotype pathway. Prokineticins are a prognostic biomarker and the correlation between hub genes and LGG requires further attention.
Collapse
|
58
|
Hu J, Zhang R, Chang Q, Ji M, Zhang H, Geng R, Li C, Wang Z. p53: A Regulator of Ferroptosis Induced by Galectin-1 Derived Peptide 3 in MH7A Cells. Front Genet 2022; 13:920273. [PMID: 35860469 PMCID: PMC9289366 DOI: 10.3389/fgene.2022.920273] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/16/2022] [Indexed: 12/21/2022] Open
Abstract
Backgrounds: Rheumatoid arthritis synovial fibroblasts (RASFs) are the primary cells responsible for destruction of marginal cartilage in rheumatoid arthritis (RA). G1dP3, a bioactive peptide derived from galectin-1 domain, possesses potent anti-inflammatory and anti-proliferation properties in RASFs. This study aimed to determine the effects of G1dP3 ferroptosis induction in RASFs and to further clarify the possible mechanisms. Methods: TNF-α was used to establish a RA model in MH7A cells. Cell Counting Kit-8 assays were employed to detect MH7A cell viability with different treatments. The occurrence of ferroptosis was examined by Lipid ROS assay, cellular labile iron pool measurement, reduced glutathione/oxidized glutathione activity, Gpx4 expression and transmission electron microscopy (TEM) morphology observation. Lentiviral-mediated siRNA interference was used to determine the downstream pathway. Results: G1dP3 markedly suppressed MH7A cell viability induced by TNF-α. G1dP3-treated MH7A cells presented the morphological features of ferroptosis. Moreover, G1dP3 triggered ferroptosis in MH7A cells by promoting the accumulation of lipid peroxides as well as iron deposition. Inhibition of ferroptosis alleviated G1dP3-mediated suppression of MH7A cell viability. Furthermore, G1dP3 increased p53 expression, which in turn transcriptionally suppressed SLC7A11, a key component of system Xc− essential for ferroptosis. Knockdown of p53 abrogated the ferroptotic effects of G1dP3 on MH7A cells. Conclusion: Our findings reveal that the bioactive peptide G1dP3 promotes RASFs ferroptosis cell death via a p53/SLC7A11 axis-dependent mechanism, suggesting its potential role in the treatment of RA.
Collapse
Affiliation(s)
- Junzheng Hu
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Rui Zhang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Qing Chang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Mingliang Ji
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Haixiang Zhang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Rui Geng
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Chao Li
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Zhen Wang
- Department of Orthopaedics, The First Hospital Affiliated to China Pharmaceutical University, Nanjing, China
- *Correspondence: Zhen Wang,
| |
Collapse
|
59
|
Ferroptosis and Its Role in Chronic Diseases. Cells 2022; 11:cells11132040. [PMID: 35805124 PMCID: PMC9265893 DOI: 10.3390/cells11132040] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 02/04/2023] Open
Abstract
Ferroptosis, which has been widely associated with many diseases, is an iron-dependent regulated cell death characterized by intracellular lipid peroxide accumulation. It exhibits morphological, biochemical, and genetic characteristics that are unique in comparison to other types of cell death. The course of ferroptosis can be accurately regulated by the metabolism of iron, lipids, amino acids, and various signal pathways. In this review, we summarize the basic characteristics of ferroptosis, its regulation, as well as the relationship between ferroptosis and chronic diseases such as cancer, nervous system diseases, metabolic diseases, and inflammatory bowel diseases. Finally, we describe the regulatory effects of food-borne active ingredients on ferroptosis.
Collapse
|
60
|
Fang J, Yuan Q, Du Z, Fei M, Zhang Q, Yang L, Wang M, Yang W, Yu J, Wu G, Hu J. Ferroptosis in brain microvascular endothelial cells mediates blood-brain barrier disruption after traumatic brain injury. Biochem Biophys Res Commun 2022; 619:34-41. [PMID: 35728282 DOI: 10.1016/j.bbrc.2022.06.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 06/12/2022] [Indexed: 11/28/2022]
Abstract
Ferroptosis is a newly recognized form of regulated cell death. Recently, growing evidence has shown that ferroptosis is involved in the pathogenesis of traumatic brain injury (TBI). However, less attention has been paid to its role in brain microvascular endothelial cells (BMVECs) and blood-brain barrier (BBB) damage, the central pathological process in secondary brain injury of TBI. Here, we established a mechanical stretch injury bEnd.3 model and a Controlled Cortical Impact (CCI) mouse model to explore the ferroptosis-related markers in brain endothelial cells after TBI in vitro and in vivo. From the results of RNA-seq analysis, RT-qPCR and immunostaining, glutathione peroxidase 4 (GPX4) downregulation, Cyclooxygenase-2 (COX-2) upregulation, and iron accumulation were observed in brain endothelial cells after TBI both in vitro and in vivo. Furthermore, we utilized Ferrostatin-1 (Fer-1), a specific inhibitor of ferroptosis, to investigate the protective effects of ferroptosis inhibition on BBB disruption and neurological deficits. From the results of immunostaining, transmission electron microscopy (TEM), and western blotting, we demonstrated that Fer-1 significantly reduced BMVECs death, BBB permeability, and tight junction loss at 3 days after TBI. The neurological tests including grid walking, rotarod test, and wire-hanging test showed that Fer-1 administration exerted neuroprotective effects in the early stage of TBI. Our findings provided evidences for inhibition of BMVECs ferroptosis as a promising therapeutic target against TBI-induced BBB disruption.
Collapse
Affiliation(s)
- Jiang Fang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Qiang Yuan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Zhuoying Du
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Maoxing Fei
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Quan Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Lei Yang
- Department of Neurosurgery & Neurocritical Care, Huashan Hospital, Fudan University, Shanghai, China
| | - Meihua Wang
- Department of Neurosurgery & Neurocritical Care, Huashan Hospital, Fudan University, Shanghai, China
| | - Weijian Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Jian Yu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Gang Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Jin Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
| |
Collapse
|
61
|
Jin Z, Liu M, Huang X, Zhang X, Qu Z, Zhu JJ, Min Q. Top-Down Rational Engineering of Heteroatom-Doped Graphene Quantum Dots for Laser Desorption/Ionization Mass Spectrometry Detection and Imaging of Small Biomolecules. Anal Chem 2022; 94:7609-7618. [PMID: 35575691 DOI: 10.1021/acs.analchem.2c00802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) is widely applied in mapping macrobiomolecules in tissues, but it is still limited in profiling low-molecular-weight (MW) compounds (typically metabolites) due to ion interference and suppression by organic matrices. Here, we present a versatile "top-down" strategy for rational engineering of carbon material-based matrices, by which heteroatom-doped graphene quantum dots (HGQDs) were manufactured for LDI MS detection and imaging of small biomolecules. The HGQDs derived from parent materials inherited the π-conjugated networks and doping sites for promoting energy transfer and negative ion generation, while their extremely small size guaranteed the matrix uniformity and signal reproducibility in LDI MSI. Compared to other HGQDs, nitrogen-doped graphene quantum dots (NGQDs) exhibited superior capability of assisting LDI of various small molecules, including amino acids, fatty acids, saccharides, small peptides, nucleobases, anticancer drugs, and bisphenol pollutants. Density functional theory simulations also corroborated that the LDI efficiency was markedly raised by the proton-capturing pyridinic nitrogen species and compromised by the electron-deficient boron dopants. NGQDs-assisted LDI MS further enabled label-free investigation on enzyme kinetics using an ordinary short peptide as the substrate. Moreover, due to the high salt tolerance and signal reproducibility, the proposed negative-ion NGQDs-assisted LDI MSI was able to reveal the abundance and distribution of low-MW species in rat brain tissue and achieved the imaging of low-MW lipids in coronally sectioned rat brains subjected to traumatic brain injury. Our work offers a new route for customizing nanomaterial matrices toward LDI MSI of small biomolecules in biomedical and pathological research.
Collapse
Affiliation(s)
- Zehui Jin
- State Key Laboratory of Analytical Chemistry for Life Sciences, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Meng Liu
- State Key Laboratory of Analytical Chemistry for Life Sciences, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiaodan Huang
- State Key Laboratory of Analytical Chemistry for Life Sciences, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xuemeng Zhang
- State Key Laboratory of Analytical Chemistry for Life Sciences, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Zexing Qu
- Institute of Theoretical Chemistry, Jilin University, Changchun 130023, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Sciences, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qianhao Min
- State Key Laboratory of Analytical Chemistry for Life Sciences, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
62
|
Liang H, Tang T, Huang H, Li T, Gao C, Han Y, Yuan B, Gao S, Wang H, Zhou ML. Peroxisome proliferator-activated receptor-γ ameliorates neuronal ferroptosis after traumatic brain injury in mice by inhibiting cyclooxygenase-2. Exp Neurol 2022; 354:114100. [PMID: 35490721 DOI: 10.1016/j.expneurol.2022.114100] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/20/2022] [Accepted: 04/24/2022] [Indexed: 01/08/2023]
Abstract
Among the multiple kinds of neuronal cell death triggered by traumatic brain injury (TBI), ferroptosis, an iron-dependent lipid peroxidative regulatory cell death, has a critical role. Peroxisome proliferator-activated receptor-γ (PPARγ) is a nuclear transcription factor that regulates lipid metabolism and suppresses neuronal inflammation. However, the role of PPARγ in neuronal ferroptosis induced by TBI remains unclear. Here, we investigated the regulatory effect of PPARγ on neuronal ferroptosis in a weight-drop TBI model in vivo and an RAS-selective lethal 3 (RSL3)-activated ferroptotic neuronal model in vitro. PPARγ was mainly localized in the nucleus of neurons and was decreased in both the in vivo TBI model and the in vitro ferroptotic neuronal model. The addition of a specific agonist, pioglitazone, activated PPARγ, which protected neuronal function post-TBI in vivo and increased the viability of ferroptotic neurons in vitro. Further investigation suggested that PPARγ probably attenuates neuronal ferroptosis by downregulating cyclooxygenase-2 (COX2) protein expression levels in vivo and in vitro. This study revealed the relationship among PPARγ, ferroptosis and TBI and identified a potential target for comprehensive TBI treatment.
Collapse
Affiliation(s)
- Hui Liang
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Ting Tang
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Hanyu Huang
- Department of Neurosurgery, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, PR China
| | - Tao Li
- Department of Neurosurgery, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, PR China
| | - Chaochao Gao
- Department of Neurosurgery, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, PR China
| | - Yanling Han
- Department of Neurosurgery, Jinling Hospital, Nanjing, PR China
| | - Bin Yuan
- Department of Neurosurgery, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, PR China
| | - Shengqing Gao
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Handong Wang
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China.
| | - Meng-Liang Zhou
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China.
| |
Collapse
|
63
|
Liu C, Sun W, Zhu T, Shi S, Zhang J, Wang J, Gao F, Ou Q, Jin C, Li J, Xu JY, Zhang J, Tian H, Xu GT, Lu L. Glia maturation factor-β induces ferroptosis by impairing chaperone-mediated autophagic degradation of ACSL4 in early diabetic retinopathy. Redox Biol 2022; 52:102292. [PMID: 35325805 PMCID: PMC8942824 DOI: 10.1016/j.redox.2022.102292] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 12/27/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the leading causes of blindness in the world, and timely prevention and treatment are very important. Previously, we found that a neurodegenerative factor, Glia maturation factor-β (GMFB), was upregulated in the vitreous at a very early stage of diabetes, which may play an important role in pathogenesis. Here, we found that in a high glucose environment, large amounts of GMFB protein can be secreted in the vitreous, which translocates the ATPase ATP6V1A from the lysosome, preventing its assembly and alkalinizing the lysosome in the retinal pigment epithelial (RPE) cells. ACSL4 protein can be recognized by HSC70, the receptor for chaperone-mediated autophagy, and finally digested in the lysosome. Abnormalities in the autophagy–lysosome degradation process lead to its accumulation, which catalyzes the production of lethal lipid species and finally induces ferroptosis in RPE cells. GMFB antibody, lysosome activator NKH477, CMA activator QX77, and ferroptosis inhibitor Liproxstatin-1 were all effective in preventing early diabetic retinopathy and maintaining normal visual function, which has powerful clinical application value. Our research broadens the understanding of the relationship between autophagy and ferroptosis and provides a new therapeutic target for the treatment of DR. Glia maturation factor-β (GMFB) was upregulated in the vitreous at a very early stage of diabetes. Extracellular GMFB can induce ferroptosis and lysosome dysfunction in retinal RPE cells. ACSL4 protein can be degraded through chaperone-mediated autophagy (CMA), and finally digested in the lysosome. Lysosome dysfunction leads to the accumulation of ACSL4, which induces ferroptosis in RPE cells.
Collapse
|
64
|
Tuo QZ, Liu Y, Xiang Z, Yan HF, Zou T, Shu Y, Ding XL, Zou JJ, Xu S, Tang F, Gong YQ, Li XL, Guo YJ, Zheng ZY, Deng AP, Yang ZZ, Li WJ, Zhang ST, Ayton S, Bush AI, Xu H, Dai L, Dong B, Lei P. Thrombin induces ACSL4-dependent ferroptosis during cerebral ischemia/reperfusion. Signal Transduct Target Ther 2022; 7:59. [PMID: 35197442 PMCID: PMC8866433 DOI: 10.1038/s41392-022-00917-z] [Citation(s) in RCA: 126] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/14/2021] [Accepted: 01/31/2022] [Indexed: 02/08/2023] Open
Abstract
Ischemic stroke represents a significant danger to human beings, especially the elderly. Interventions are only available to remove the clot, and the mechanism of neuronal death during ischemic stroke is still in debate. Ferroptosis is increasingly appreciated as a mechanism of cell death after ischemia in various organs. Here we report that the serine protease, thrombin, instigates ferroptotic signaling by promoting arachidonic acid mobilization and subsequent esterification by the ferroptotic gene, acyl-CoA synthetase long-chain family member 4 (ACSL4). An unbiased multi-omics approach identified thrombin and ACSL4 genes/proteins, and their pro-ferroptotic phosphatidylethanolamine lipid products, as prominently altered upon the middle cerebral artery occlusion in rodents. Genetically or pharmacologically inhibiting multiple points in this pathway attenuated outcomes of models of ischemia in vitro and in vivo. Therefore, the thrombin-ACSL4 axis may be a key therapeutic target to ameliorate ferroptotic neuronal injury during ischemic stroke.
Collapse
Affiliation(s)
- Qing-Zhang Tuo
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Yu Liu
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Zheng Xiang
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Hong-Fa Yan
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Ting Zou
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Yang Shu
- Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Xu-Long Ding
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Jin-Jun Zou
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Shuo Xu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Fei Tang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Yan-Qiu Gong
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Xiao-Lan Li
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Yu-Jie Guo
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Zhao-Yue Zheng
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Ai-Ping Deng
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Zhang-Zhong Yang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Wen-Jing Li
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Shu-Ting Zhang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Heng Xu
- Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Lunzhi Dai
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China.
| | - Biao Dong
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China.
| | - Peng Lei
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China. .,Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 610041, Chengdu, Sichuan, China.
| |
Collapse
|
65
|
The Neuronal Transcription Factor Creb3l1 Potential Upregulates Ntrk2 in the Hypertensive Microenvironment to Promote Vascular Smooth Muscle Cell-Neuron Interaction and Prevent Neurons from Ferroptosis: A Bioinformatic Research of scRNA-seq Data. DISEASE MARKERS 2022; 2022:8339759. [PMID: 35186168 PMCID: PMC8853773 DOI: 10.1155/2022/8339759] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022]
Abstract
Background There is still a lack of knowledge regarding the association between hypertension and ferroptosis. A single-cell approach was used to study the changes in neuropeptide expression as they might contribute to the mechanisms leading to ferroptosis in a hypertensive microenvironment. Methods We analyzed 11798 cells from the SHR group and 12589 cells from the WKY group of mouse arterial cells. CellPhoneDB was used for cell communication analysis, and the SCENIC method was used to identify key transcription factors in neurons. The correlation between Ntrk2 and ferroptosis-related genes was further analyzed and validated via quantitative polymerase chain reaction. Results The arterial cells were clustered into six cell types. Ligand-receptor analysis suggested that Ngf, Ntf3, Cxcr4, and Ntrk2 were key neuropeptide-related genes involved in the communication between vascular smooth muscle cells and neural cells. In the hypertensive microenvironment, the neuronal transcription factor Creb3l1 appears to play a key role in the upregulation of Ntrk2 to promote the interaction between neurons and vascular smooth muscle cells. An association between Ntrk2 and the ferroptosis death inhibitor Gpx4 was suggested. RT-qPCR experiments confirmed that Ntrk2 downregulation in neural cells was followed by downregulated expression of Gpx4. Conclusions Creb3l1, a key transcription factor in vascular neurons, may upregulate Ntrk2 to promote vascular smooth muscle cell-neuron interaction and thereby potentially prevent ferroptosis in neurons.
Collapse
|
66
|
Lattanzi R, Miele R. Versatile Role of Prokineticins and Prokineticin Receptors in Neuroinflammation. Biomedicines 2021; 9:1648. [PMID: 34829877 PMCID: PMC8615546 DOI: 10.3390/biomedicines9111648] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 01/15/2023] Open
Abstract
Prokineticins are a new class of chemokine-like peptides involved in a wide range of biological and pathological activities. In particular, prokineticin 2 (PK2), prokineticin receptor 1 (PKR1) and prokineticin receptor 2 (PKR2) play a central role in modulating neuroinflammatory processes. PK2 and PKRs, which are physiologically expressed at very low levels, are strongly upregulated during inflammation and regulate neuronal-glial interaction. PKR2 is mainly overexpressed in neurons, whereas PKR1 and PK2 are mainly overexpressed in astrocytes. Once PK2 is released in inflamed tissue, it is involved in both innate and adaptive responses: it triggers macrophage recruitment, production of pro-inflammatory cytokines, and reduction of anti-inflammatory cytokines. Moreover, it modulates the function of T cells through the activation of PKR1 and directs them towards a pro-inflammatory Th1 phenotype. Since the prokineticin system appears to be upregulated following a series of pathological insults leading to neuroinflammation, we will focus here on the involvement of PK2 and PKRs in those pathologies that have a strong underlying inflammatory component, such as: inflammatory and neuropathic pain, Alzheimer's disease, Parkinson's disease, multiple sclerosis, stroke, obesity, diabetes, and gastrointestinal inflammation.
Collapse
Affiliation(s)
- Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Department of Biochemical Sciences “A. Rossi Fanelli”, CNR Institute of Molecular Biology and Pathology, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| |
Collapse
|