1
|
Shen L, Tian Q, Ran Q, Gan Q, Hu Y, Du D, Qin Z, Duan X, Zhu X, Huang W. Z-Ligustilide: A Potential Therapeutic Agent for Atherosclerosis Complicating Cerebrovascular Disease. Biomolecules 2024; 14:1623. [PMID: 39766330 PMCID: PMC11726876 DOI: 10.3390/biom14121623] [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: 11/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Atherosclerosis (AS) is one of the major catalysts of ischemic cerebrovascular disease, and the death and disease burden from AS and its cerebrovascular complications are increasing. Z-ligustilide (Z-LIG) is a key active ingredient in Angelica sinensis (Oliv.) Diels and Ligusticum chuanxiong Hort. In this paper, we first introduced LIG's physicochemical properties and pharmacokinetics. Then, we reviewed Z-LIG's intervention and therapeutic mechanisms on AS and its cerebrovascular complications. The mechanisms of Z-LIG intervention in AS include improving lipid metabolism, antioxidant and anti-inflammatory effects, protecting vascular endothelium, and inhibiting vascular endothelial fibrosis, pathological thickening, and plaque calcification. In ischemic cerebrovascular diseases complicated by AS, Z-LIG exerts practical neuroprotective effects in ischemic stroke (IS), transient ischemic attack (TIA), and vascular dementia (VaD) through anti-neuroinflammatory, anti-oxidation, anti-neuronal apoptosis, protection of the blood-brain barrier, promotion of mitochondrial division and angiogenesis, improvement of cholinergic activity, inhibition of astrocyte proliferation, and endoplasmic reticulum stress. This paper aims to provide a basis for subsequent studies of Z-LIG in the prevention and treatment of AS and its cerebrovascular complications and, thus, to promote the development of interventional drugs for AS.
Collapse
Affiliation(s)
- Longyu Shen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianqian Tian
- Faculty of Social Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Qiqi Ran
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianrong Gan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Donglian Du
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Zehua Qin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyi Duan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyun Zhu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Wei Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| |
Collapse
|
2
|
Ma J, Guo Q, Shen MQ, Li W, Zhong QX, Qian ZM. Apolipoprotein E is required for brain iron homeostasis in mice. Redox Biol 2023; 64:102779. [PMID: 37339558 PMCID: PMC10363452 DOI: 10.1016/j.redox.2023.102779] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Apolipoprotein E deficiency (ApoE-/-) increases progressively iron in the liver, spleen and aortic tissues with age in mice. However, it is unknown whether ApoE affects brain iron. METHODS We investigated iron contents, expression of transferrin receptor 1 (TfR1), ferroportin 1 (Fpn1), iron regulatory proteins (IRPs), aconitase, hepcidin, Aβ42, MAP2, reactive oxygen species (ROS), cytokines and glutathione peroxidase 4 (Gpx4) in the brain of ApoE-/- mice. RESULTS We demonstrated that ApoE-/- induced a significant increase in iron, TfR1 and IRPs and a reduction in Fpn1, aconitase and hepcidin in the hippocampus and basal ganglia. We also showed that replenishment of ApoE absent partly reversed the iron-related phenotype in ApoE-/- mice at 24-months old. In addition, ApoE-/- induced a significant increase in Aβ42, MDA, 8-isoprostane, IL-1β, IL-6, and TNFα and a reduction in MAP2 and Gpx4 in hippocampus, basal ganglia and/or cortex of mice at 24-months old. CONCLUSIONS Our findings implied that ApoE is required for brain iron homeostasis and ApoE-/--induced increase in brain iron is due to the increased IRP/TfR1-mediated cell-iron uptake as well as the reduced IRP/Fpn1 associated cell-iron export and suggested that ApoE-/- induced neuronal injury resulted mainly from the increased iron and subsequently ROS, inflammation and ferroptosis.
Collapse
Affiliation(s)
- Juan Ma
- Department of Neurology, Affiliated Hospital, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226001, China; Laboratory of Neuropharmacology of Pharmacy School, and National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China.
| | - Qian Guo
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, 881 Yonghe Road, Nantong, Jiangsu, 226001, China; Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai, 200444, China.
| | - Meng-Qi Shen
- Department of Neurology, Affiliated Hospital, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226001, China.
| | - Wei Li
- Department of Neurology, Affiliated Hospital, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226001, China.
| | - Qi-Xin Zhong
- Department of Cardiovascular Medicine, Shenzhen Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518034, China.
| | - Zhong-Ming Qian
- Department of Neurology, Affiliated Hospital, and Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
3
|
Jiang L, Shen M, Bao Y, Qian Z. Verapamil downregulates iron uptake and upregulates divalent metal transporter 1 expression in H9C2 cardiomyocytes. Fundam Clin Pharmacol 2022; 36:985-991. [DOI: 10.1111/fcp.12793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 03/30/2022] [Accepted: 04/28/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Li‐Rong Jiang
- Laboratory of Neuropharmacology Fudan University School of Pharmacy Shanghai China
| | - Meng‐Qi Shen
- Institute of Translational and Precision Medicine Nantong University Nantong China
| | - Yu‐Xin Bao
- Research Center for Medicine and Biology Zunyi Medical University Zunyi China
| | - Zhong‐Ming Qian
- Laboratory of Neuropharmacology Fudan University School of Pharmacy Shanghai China
- Institute of Translational and Precision Medicine Nantong University Nantong China
| |
Collapse
|
4
|
Wu XM, Qian C, Jiang F, Bao YX, Qian ZM, Ke Y. The involvement of nuclear factor-κB in astroprotection against ischemia-reperfusion injury by ischemia-preconditioned neurons. J Cell Physiol 2021; 236:4515-4527. [PMID: 33442879 DOI: 10.1002/jcp.30168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/21/2020] [Accepted: 11/06/2020] [Indexed: 11/12/2022]
Abstract
Ischemic preconditioned (IP) neurons protect astrocytes against ischemia/reperfusion (I/R)-induced injury by inhibiting oxidative stress. However, the relevant mechanisms are unknown. Based on the role of nuclear factor-κB (NF-κB) in cell survival and adaption to oxidative stress, we hypothesized that NF-κB might be associated with astroprotection induced by IP neurons via upregulation of antioxidant enzymes. Here, we investigated the effects of IP neurons on NF-κB activation, cell viability, reactive oxygen species (ROS), expression of antioxidant enzymes, erythropoietin (EPO), and tumor necrosis factor α (TNF-α), in the presence or absence of BAY11-7082 (an NF-κB inhibitor), anti-EPO, and anti-TNF-α antibodies, in astrocytes treated with or without I/R. We found that IP neurons could keep NF-κB activation at a relatively higher but beneficial level, and in turn, upregulated the activity of antioxidant enzymes and hence enhanced cell viability and reduced ROS in I/R treated astrocytes. The results collectively indicated that IP neurons are able to significantly inhibit the I/R-induced NF-κB overactivation, probably via EPO and TNF-α, being essential for IP neuron-induced astroprotection under the conditions of I/R. We concluded that NF-κB-mediated antioxidative stress is one of the mechanisms by which IP neurons protect astrocytes against I/R injury.
Collapse
Affiliation(s)
- Xiao-Mei Wu
- Institute of Translational & Precision Medicine and Institute for Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Fei Jiang
- Institute of Translational & Precision Medicine and Institute for Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Yu-Xin Bao
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Zhong-Ming Qian
- Institute of Translational & Precision Medicine and Institute for Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
- Laboratory of Neuropharmacology, School of Pharmacy & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| |
Collapse
|
5
|
Jurado M, Castaño Ó, Zorzano A. Stochastic modulation evidences a transitory EGF-Ras-ERK MAPK activity induced by PRMT5. Comput Biol Med 2021; 133:104339. [PMID: 33910125 DOI: 10.1016/j.compbiomed.2021.104339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
The extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) pathway involves a three-step cascade of kinases that transduce signals and promote processes such as cell growth, development, and apoptosis. An aberrant response of this pathway is related to the proliferation of cell diseases and tumors. By using simulation modeling, we document that the protein arginine methyltransferase 5 (PRMT5) modulates the MAPK pathway and thus avoids an aberrant behavior. PRMT5 methylates the Raf kinase, reducing its catalytic activity and thereby, reducing the activation of ERK in time and amplitude. Two minimal computational models of the epidermal growth factor (EGF)-Ras-ERK MAPK pathway influenced by PRMT5 were proposed: a first model in which PRMT5 is activated by EGF and a second one in which PRMT5 is stimulated by the cascade response. The reported results show that PRMT5 reduces the time duration and the expression of the activated ERK in both cases, but only in the first model PRMT5 limits the EGF range that generates an ERK activation. Based on our data, we propose the protein PRMT5 as a regulatory factor to develop strategies to fight against an excessive activity of the MAPK pathway, which could be of use in chronic diseases and cancer.
Collapse
Affiliation(s)
- Manuel Jurado
- Biotechnology Ph.D. Programme, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Óscar Castaño
- Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain; CIBER in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain; Bioelectronics Unit and Nanobioengineering Lab., Institute for Nanoscience and Nanotechnology of the University of Barcelona (IN2UB), Barcelona, Spain.
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; CIBER of Diabetes and Associated Metabolic Diseases, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Hepatocyte growth factor protects PC12 cells against OGD/R-induced injury by reducing iron. Biosci Rep 2021; 40:222408. [PMID: 32186328 PMCID: PMC7109004 DOI: 10.1042/bsr20200287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/11/2020] [Accepted: 03/16/2020] [Indexed: 01/20/2023] Open
Abstract
In the light of hepatocyte growth factor (HGF) the inhibiting role on the expression of hepcidin, we hypothesized that HGF might be able to reduce cell and tissue iron by increasing ferroportin 1 (Fpn1) content and Fpn1-mediated iron release from cells and tissues. The hypothesized ability of HGF to reduce iron might be one of the mechanisms associated with its neuroprotective action under the conditions of ischemia/reperfusion (I/R). Here, we investigated the effects of HGF on the expression of hepcidin as well as transferrin receptor 1 (TfR1), divalent metal transporter 1 (DMT1), Fpn1, ferritin and iron regulatory proteins (IRPs) in oxygen-glucose deprivation and reoxygenation (OGD/R)-treated PC12 cells by real-time PCR and Western blot analysis. We demonstrated that HGF could completely reverse the OGD/R-induced reduction in Fpn1 and IRP1 expression and increase in ferritin light chain protein and hepcidin mRNA levels in PC12 cells. It was concluded that HGF protects PC12 cells against OGD/R-induced injury mainly by reducing cell iron contents via the up-regulation of Fpn1 and increased Fpn1-mediated iron export from cells. Our findings suggested that HGF may also be able to ameliorate OGD/R or I/R-induced overloading of brain iron by promoting Fpn1 expression.
Collapse
|
7
|
Hormesis and Ginkgo biloba (GB): Numerous biological effects of GB are mediated via hormesis. Ageing Res Rev 2020; 64:101019. [PMID: 31931153 DOI: 10.1016/j.arr.2020.101019] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/27/2019] [Accepted: 01/09/2020] [Indexed: 01/11/2023]
Abstract
Ginkgo biloba (GB) extracts have been shown to commonly induce biphasic dose responses in a range of cell types and endpoints (e.g., cochlea neural stem cells, cell viability, cell proliferation). The magnitude and width of the low dose stimulation of these biphasic dose responses are similar to those reported for hormetic dose responses. These hormetic dose responses occur within direct stimulatory responses as well as in preconditioning experimental protocols, displaying acquired resistance within an adaptive homeodynamic and temporal framework and repeated measurement protocols. The demonstrated GB dose responses further reflect the general occurrence of hormetic dose responses that consistently appear to be independent of the biological model, endpoint, inducing agent, and/or mechanism. These findings have important implications for consideration(s) of study designs involving dose selection, dose spacing, sample size, and statistical power. This illustrates and strengthens the need to characterize the low dose stimulatory response range and optimal dose in order to explore potential public health and clinical applications of plant-derived agents, such as GB.
Collapse
|
8
|
Labkovich M, Jacobs EB, Bhargava S, Pasquale LR, Ritch R. Ginkgo Biloba Extract in Ophthalmic and Systemic Disease, With a Focus on Normal-Tension Glaucoma. Asia Pac J Ophthalmol (Phila) 2020; 9:215-225. [PMID: 32282348 PMCID: PMC7299225 DOI: 10.1097/apo.0000000000000279] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 02/05/2020] [Indexed: 12/15/2022] Open
Abstract
Glaucoma is a neurodegenerative eye disease that results in retinal ganglion cell loss and ultimately loss of vision. Elevated intraocular pressure (IOP) is the most common known risk factor for retinal ganglion cell damage and visual field loss, and the only modifiable risk factor proven to reduce the development and progression of glaucoma. This has greatly influenced our approach and assessment in terms of diagnosis and treatment. However, as many as ≥50% of patients with progressive vision loss from primary open angle glaucoma without IOP elevation (≤22 mm Hg) have been reported in the United States and Canada; 90% in Japan and 80% in Korea. Extensive research is currently underway to identify the etiology of risk factors for glaucoma other than or in addition to elevated IOP (so-called "normal-tension" glaucoma; NTG) and use this knowledge to expand available treatment options. Currently, Food and Drug Administration-approved medications for glaucoma exclusively target elevated IOP, suggesting the need for additional approaches to treatment options beyond the current scope as the definition of glaucoma changes to encompass cellular and molecular mechanisms. This review focuses on alternative medical approaches, specifically Ginkgo Biloba extract, as a potential treatment option for normal-tension glaucoma.
Collapse
Affiliation(s)
- Margarita Labkovich
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY
| | - Erica B. Jacobs
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY
| | - Siddharth Bhargava
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY
| | - Louis R. Pasquale
- Department of Ophthalmology, Eye and Vision Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert Ritch
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY
| |
Collapse
|
9
|
Chen YJ, Qian ZM, Sheng Y, Zheng J, Liu Y. Angiotensin II down-regulates transferrin receptor 1 and ferroportin 1 expression in Neuro-2a cells via activation of type-1 receptor. Neurosci Lett 2019; 716:134684. [PMID: 31830506 DOI: 10.1016/j.neulet.2019.134684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/30/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
Angiotensin II (ANGII) modulates expression of iron intake and export proteins in cultured neurons. However, the relevant mechanisms have not been fully elucidated. Here, we investigated the effects of ANGII and/or candesartan, a ANGII-Type-1 Receptor (AT1R) antagonist, and PD123319, a ANGII-Type-2 Receptor (AT2R) antagonist on expression of transferrin receptor 1 (TfR1), ferroportin 1 (Fpn1)and ferritin as well as iron regulatory proteins (IRPs), hepcidin and nuclear factor E2-related factor 2 (Nrf2) in Neuro-2a cells. We demonstrated that ANGII induces a significant reduction in expression of TfR1, Fpn1, IRP2 proteins and Nrf2 mRNA and an increase in ferritin protein and hepcidin mRNA, while candesartan, but not PD123319, significantly attenuated or reversed all these ANGII-induced changes in Neuro-2a cells. These findings imply that ANGII down-regulates TfR1 expression likely via the AT1R/IRP2 pathway, and Fpn1 expression via ATR1/hepcidin and AT1R/ Nrf2 pathways.
Collapse
Affiliation(s)
- Yun-Jin Chen
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institute of Translational & Precision Medicine, Nantong University, Nantong, JS, 226019, China.
| | - Yuan Sheng
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jie Zheng
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yong Liu
- Department of Neurology, The Second Affiliated (Xinqiao) Hospital, The Army (Third Military) Medical University, Chongqing, China.
| |
Collapse
|
10
|
Huang SN, Ruan HZ, Chen MYJ, Zhou G, Qian ZM. Aspirin increases ferroportin 1 expression by inhibiting hepcidin via the JAK/STAT3 pathway in interleukin 6-treated PC-12 cells. Neurosci Lett 2018; 662:1-5. [DOI: 10.1016/j.neulet.2017.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/16/2017] [Accepted: 10/02/2017] [Indexed: 12/31/2022]
|
11
|
Zhou YF, Zhang C, Yang G, Qian ZM, Zhang MW, Ma J, Zhang FL, Ke Y. Hepcidin Protects Neuron from Hemin-Mediated Injury by Reducing Iron. Front Physiol 2017; 8:332. [PMID: 28588503 PMCID: PMC5440571 DOI: 10.3389/fphys.2017.00332] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/08/2017] [Indexed: 01/05/2023] Open
Abstract
Hemin plays a key role in mediating secondary neuronal injury after intracerebral hemorrhage (ICH) and the cell toxicity of hemin is thought to be due to iron that is liberated when hemin is degraded. In a recent study, we demonstrated the iron regulatory hormone hepcidin reduces brain iron in iron-overloaded rats. Therefore, we hypothesized that hepcidin might be able to reduce iron and then protect neurons from hemin or iron-mediated neurotoxicity in hemin-treated neuronal cells. Here, we tested the hypothesis and demonstrated that ad-hepcidin and hepcidin peptide both have the ability to suppress the hemin-induced increase in LDH release and apoptotic cell numbers, to reduce cell iron and ferritin contents, and to inhibit expression of transferrin receptor 1, divalent metal transporter 1, and ferroportin 1 in hemin-treated neurons. We conclude that hepcidin protects neuron from hemin-mediated injury by reducing iron via inhibition of expression of iron transport proteins.
Collapse
Affiliation(s)
- Yu-Fu Zhou
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China
| | - Chao Zhang
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| | - Guang Yang
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China
| | - Meng-Wan Zhang
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| | - Juan Ma
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| | - Fa-Li Zhang
- Laboratory of Neuropharmacology, School of Pharmacy, Fudan UniversityShanghai, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| | - Ya Ke
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong KongShatin, Hong Kong
| |
Collapse
|
12
|
Wu XM, Qian C, Zhou YF, Yan YC, Luo QQ, Yung WH, Zhang FL, Jiang LR, Qian ZM, Ke Y. Bi-directionally protective communication between neurons and astrocytes under ischemia. Redox Biol 2017; 13:20-31. [PMID: 28551085 PMCID: PMC5447396 DOI: 10.1016/j.redox.2017.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/16/2017] [Accepted: 05/19/2017] [Indexed: 01/10/2023] Open
Abstract
The extensive existing knowledge on bi-directional communication between astrocytes and neurons led us to hypothesize that not only ischemia-preconditioned (IP) astrocytes can protect neurons but also IP neurons protect astrocytes from lethal ischemic injury. Here, we demonstrated for the first time that neurons have a significant role in protecting astrocytes from ischemic injury. The cultured medium from IP neurons (IPcNCM) induced a remarkable reduction in LDH and an increase in cell viability in ischemic astrocytes in vitro. Selective neuronal loss by kainic acid injection induced a significant increase in apoptotic astrocyte numbers in the brain of ischemic rats in vivo. Furthermore, TUNEL analysis, DNA ladder assay, and the measurements of ROS, GSH, pro- and anti-apoptotic factors, anti-oxidant enzymes and signal molecules in vitro and/or in vivo demonstrated that IP neurons protect astrocytes by an EPO-mediated inhibition of pro-apoptotic signals, activation of anti-apoptotic proteins via the P13K/ERK/STAT5 pathways and activation of anti-oxidant proteins via up-regulation of anti-oxidant enzymes. We demonstrated the existence of astro-protection by IP neurons under ischemia and proposed that the bi-directionally protective communications between cells might be a common activity in the brain or peripheral organs under most if not all pathological conditions.
Collapse
Affiliation(s)
- Xiao-Mei Wu
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, NT, Hong Kong, China; Department of Biochemistry, Institute for Nautical Medicine, Nantong University, Nantong 226001, China
| | - Christopher Qian
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Yu-Fu Zhou
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, NT, Hong Kong, China; Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Shanghai 201203, China
| | - Yick-Chun Yan
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Qian-Qian Luo
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Shanghai 201203, China; Department of Biochemistry, Institute for Nautical Medicine, Nantong University, Nantong 226001, China
| | - Wing-Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Fa-Li Zhang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Shanghai 201203, China
| | - Li-Rong Jiang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Shanghai 201203, China
| | - Zhong Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Shanghai 201203, China.
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| |
Collapse
|
13
|
Calabrese EJ, Calabrese V, Giordano J. The role of hormesis in the functional performance and protection of neural systems. Brain Circ 2017; 3:1-13. [PMID: 30276298 PMCID: PMC6126232 DOI: 10.4103/2394-8108.203257] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/06/2016] [Accepted: 12/13/2016] [Indexed: 02/03/2023] Open
Abstract
This paper addresses how hormesis, a biphasic dose response, can protect and affect performance of neural systems. Particular attention is directed to the potential role of hormesis in mitigating age-related neurodegenerative diseases, genetically based neurological diseases, as well as stroke, traumatic brain injury, seizure, and stress-related conditions. The hormetic dose response is of particular significance since it mediates the magnitude and range of neuroprotective processes. Consideration of hormetic dose-response concepts can also enhance the quality of study designs, including sample size/statistical power strategies, selection of treatment groups, dose spacing, and temporal/repeat measures’ features.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Viale Andrea Doria, Catania, Italy
| | - James Giordano
- Department of Neurology and Biochemistry, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
14
|
Yu SS, Jiang LR, Ling Y, Qian ZM, Zhou YF, Li J, Ke Y. Nifedipine Increases Iron Content in WKPT-0293 Cl.2 Cells via Up-Regulating Iron Influx Proteins. Front Pharmacol 2017; 8:60. [PMID: 28243203 PMCID: PMC5303744 DOI: 10.3389/fphar.2017.00060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/30/2017] [Indexed: 12/25/2022] Open
Abstract
Nifedipine was reported to enhance urinary iron excretion in iron overloaded mice. However, it remains unknown how nifedipine stimulates urinary iron excretion in the kidney. We speculated that nifedipine might inhibit the TfR1/ DMT1 (transferrin receptor 1/divalent metal transporter1)-mediated iron uptake by proximal tubule cells in addition to blocking L-type Ca2+ channels, leading to an increase in iron in lumen-fluid and then urinary iron excretion. To test this hypothesis, we investigated the effects of nifedipine on iron content and expression of TfR1, DMT1 and ferroportin1 (Fpn1) in WKPT-0293 Cl.2 cells of the S1 segment of the proximal tubule in rats, using a graphite furnace atomic absorption spectrophotometer and Western blot analysis, respectively. We demonstrated for the first time that nifedipine significantly enhanced iron content as well as TfR1 and DMT1 expression and had no effect on Fpn1 levels in the cells. We also found that ferric ammonium citrate decreased TfR1 levels, increased Fpn1 expression and had no effect on DMT1 content, while co-treatment with nifedipine and FAC increase TfR1 and DMT1 expression and also had no effect on Fpn1 levels. These findings suggest that the nifedipine-induced increase in cell iron may mainly be due to the corresponding increase in TfR1 and DMT1 expression and also imply that the effects of nifedipine on iron transport in proximal tubule cells can not explain the increase in urinary iron excretion.
Collapse
Affiliation(s)
- Shuang-Shuang Yu
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Li-Rong Jiang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Yan Ling
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Yu-Fu Zhou
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Juan Li
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy Pudong, China
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong Hong Kong, Hong Kong
| |
Collapse
|
15
|
Li WY, Li FM, Zhou YF, Wen ZM, Ma J, Ya K, Qian ZM. Aspirin down Regulates Hepcidin by Inhibiting NF-κB and IL6/JAK2/STAT3 Pathways in BV-2 Microglial Cells Treated with Lipopolysaccharide. Int J Mol Sci 2016; 17:ijms17121921. [PMID: 27999284 PMCID: PMC5187761 DOI: 10.3390/ijms17121921] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 10/26/2016] [Accepted: 11/09/2016] [Indexed: 12/16/2022] Open
Abstract
Aspirin down regulates transferrin receptor 1 (TfR1) and up regulates ferroportin 1 (Fpn1) and ferritin expression in BV-2 microglial cells treated without lipopolysaccharides (LPS), as well as down regulates hepcidin and interleukin 6 (IL-6) in cells treated with LPS. However, the relevant mechanisms are unknown. Here, we investigate the effects of aspirin on expression of hepcidin and iron regulatory protein 1 (IRP1), phosphorylation of Janus kinase 2 (JAK2), signal transducer and activator of transcription 3 (STAT3) and P65 (nuclear factor-κB), and the production of nitric oxide (NO) in BV-2 microglial cells treated with and without LPS. We demonstrated that aspirin inhibited hepcidin mRNA as well as NO production in cells treated with LPS, but not in cells without LPS, suppresses IL-6, JAK2, STAT3, and P65 (nuclear factor-κB) phosphorylation and has no effect on IRP1 in cells treated with or without LPS. These findings provide evidence that aspirin down regulates hepcidin by inhibiting IL6/JAK2/STAT3 and P65 (nuclear factor-κB) pathways in the cells under inflammatory conditions, and imply that an aspirin-induced reduction in TfR1 and an increase in ferritin are not associated with IRP1 and NO.
Collapse
Affiliation(s)
- Wan-Ying Li
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai 201203, China.
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| | - Fei-Mi Li
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai 201203, China.
| | - Yu-Fu Zhou
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai 201203, China.
| | - Zhong-Min Wen
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| | - Juan Ma
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai 201203, China.
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China.
| | - Ke Ya
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China.
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai 201203, China.
| |
Collapse
|
16
|
Luo QQ, Qian ZM, Zhou YF, Zhang MW, Wang D, Zhu L, Ke Y. Expression of Iron Regulatory Protein 1 Is Regulated not only by HIF-1 but also pCREB under Hypoxia. Int J Biol Sci 2016; 12:1191-1202. [PMID: 27766034 PMCID: PMC5069441 DOI: 10.7150/ijbs.16437] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/18/2016] [Indexed: 02/06/2023] Open
Abstract
The inconsistent of responses of IRP1 and HIF-1 alpha to hypoxia and the similar tendencies in the changes of IRP1 and pCREB contents led us to hypothesize that pCREB might be involved in the regulation of IRP1 under hypoxia. Here, we investigated the role of pCREB in IRP1 expression in HepG2 cells under hypoxia using quantitative PCR, western blot, immunofluorescence, electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP). We demonstrated that 1) Hypoxia increased pCREB levels inside of the nucleus; 2) Putative CREs were found in the IRP1 gene; 3) Nuclear extracts of HepG2 cells treated with hypoxia could bind to CRE1 and CRE3, and 100-fold competitor of putative CREs could abolish the binding activity to varying degrees; 4) pCREB was found in the CRE1 and CRE3 DNA-protein complexes of EMSA; 5) CRE1 and CRE3 binding activity of IRP1 depended on CREB activation but not on HIF-1; 6) Increased IRP1 expression under hypoxia could be prevented by LY294002; 7) ChIP assays demonstrated that pCREB binds to IRP1 promoter; and 8) HIF-1 and/or HIF-2 siRNA had no effect on the expression of pCREB and IRP1 proteins in cells treated with hypoxia for 8 hours. Our findings evidenced for the involvement of pCREB in IRP1 expression and revealed a dominant role of PI3K/Akt pathway in CREB activation under hypoxia and also suggested that dual-regulation of IRP1 expression by HIF-1 and pCERB or other transcription factor(s) under hypoxia might be a common mechanism in most if not all of hypoxia-inducible genes.
Collapse
Affiliation(s)
- Qian-Qian Luo
- Department of Biochemistry, Institute for Nautical Medicine, Nantong University, Nantong, 226001, China; Laboratory of Neuropharmacology, FudanUniversity School of Pharmacy,826 Zhang Heng Road, Pu Dong, Shanghai201203, China
| | - Zhong-Ming Qian
- Department of Biochemistry, Institute for Nautical Medicine, Nantong University, Nantong, 226001, China; Laboratory of Neuropharmacology, FudanUniversity School of Pharmacy,826 Zhang Heng Road, Pu Dong, Shanghai201203, China
| | - Yu-Fu Zhou
- Laboratory of Neuropharmacology, FudanUniversity School of Pharmacy,826 Zhang Heng Road, Pu Dong, Shanghai201203, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, NT, Hong Kong
| | - Meng-Wan Zhang
- Laboratory of Neuropharmacology, FudanUniversity School of Pharmacy,826 Zhang Heng Road, Pu Dong, Shanghai201203, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, NT, Hong Kong
| | - Dang Wang
- Department of Biochemistry, Institute for Nautical Medicine, Nantong University, Nantong, 226001, China
| | - Li Zhu
- Department of Biochemistry, Institute for Nautical Medicine, Nantong University, Nantong, 226001, China
| | - Ya Ke
- Laboratory of Neuropharmacology, FudanUniversity School of Pharmacy,826 Zhang Heng Road, Pu Dong, Shanghai201203, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, NT, Hong Kong
| |
Collapse
|
17
|
Chen P, Li FM, Zhou YF, Qian C, Li J, Jiang LR, Qian ZM. Effects of alpha-lipoic acid on expression of iron transport and storage proteins in BV-2 microglia cells. Pharmacol Rep 2016; 69:1-5. [PMID: 27755990 DOI: 10.1016/j.pharep.2016.09.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/29/2016] [Accepted: 09/09/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND The antioxidant properties of alpha-lipoic acid (ALA) are associated with its ability to reduce iron in cells and tissues, which is partly due to its inhibiting effect on iron uptake from transferrin and its promoting effect on iron deposition into ferritin. However, the relevant mechanisms are unknown. METHODS We therefore investigated the effects of ALA on the expression of transferrin receptor 1 (TfR1), divalent metal transporter 1 (DMT1), ferroportin 1 (Fpn1) and ferritin in BV-2 microglia cells. RESULTS We demonstrated that ALA significantly inhibited DMT1 expression, lowered ferritin-light-chain (Ft-L) and ferritin-heavy-chain (Ft-H) content, and had no effect on TfR1 and Fpn1 in BV-2 microglia cells. This indicated that the inhibiting effect of ALA on DMT1 might be one of the causes of the ALA-induced reduction in cellular transferrin-bound-iron uptake. We also demonstrated that ALA enhanced DMT1 and TfR1 expression in ferric ammonium citrate (FAC)-treated cells. FAC treatment led to a significant increase in Ft-L, Ft-H and Fpn1, and pre-treatment with ALA resulted in a further increase in the contents of Ft-L and Ft-H but not Fpn1 in cells. CONCLUSIONS ALA could up-regulate TfR1, DMT1 and ferritin expression when iron is increased outside of the cell, promoting iron deposition into ferritin by increasing cell iron uptake, and then reducing free iron both inside and outside of the cell.
Collapse
Affiliation(s)
- Ping Chen
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Pu Dong, Shanghai 201203, China
| | - Fei-Mi Li
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Pu Dong, Shanghai 201203, China
| | - Yu-Fu Zhou
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Pu Dong, Shanghai 201203, China
| | - Christopher Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Pu Dong, Shanghai 201203, China
| | - Juan Li
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Pu Dong, Shanghai 201203, China
| | - Li-Rong Jiang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Pu Dong, Shanghai 201203, China
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, 826 Zhang Heng Road, Pu Dong, Shanghai 201203, China.
| |
Collapse
|
18
|
Preconditioning is hormesis part I: Documentation, dose-response features and mechanistic foundations. Pharmacol Res 2016; 110:242-264. [DOI: 10.1016/j.phrs.2015.12.021] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/16/2022]
|
19
|
Preconditioning is hormesis part II: How the conditioning dose mediates protection: Dose optimization within temporal and mechanistic frameworks. Pharmacol Res 2016; 110:265-275. [DOI: 10.1016/j.phrs.2015.12.020] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 01/02/2023]
|
20
|
Effects of aspirin on expression of iron transport and storage proteins in BV-2 microglial cells. Neurochem Int 2015; 91:72-7. [DOI: 10.1016/j.neuint.2015.10.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/24/2015] [Accepted: 10/26/2015] [Indexed: 11/23/2022]
|
21
|
Wu XM, Qian ZM, Zhu L, Du F, Yung WH, Gong Q, Ke Y. Neuroprotective effect of ligustilide against ischaemia-reperfusion injury via up-regulation of erythropoietin and down-regulation of RTP801. Br J Pharmacol 2012; 164:332-43. [PMID: 21410687 DOI: 10.1111/j.1476-5381.2011.01337.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Ligustilide, the main lipophilic component of Danggui, has been reported to protect the brain against ischaemic injury. However, the mechanisms are unknown. Here, we investigated the roles of erythropoietin (EPO) and the stress-induced protein RTP801 in neuroprotection provided by ligustilide against ischaemia-reperfusion (I/R) damage to the brain. EXPERIMENTAL APPROACH The efficacy of ligustilide against I/R damage was assessed by neurological deficit, infarct volume and cell viability, using the middle cerebral artery occlusion model in rats in vivo and rat cultured neurons in vitro. EPO and RTP801 were analysed by Western blot. Over-expression of RTP801 was achieved by transfection of an expression plasmid. KEY RESULTS Ligustilide decreased the neurological deficit score, infarct volume and RTP801 expression and increased EPO transcription in I/R rats, and increased cell viability and EPO and decreased LDH release and RTP801 in I/R neurons. Also, ligustilide increased ERK phosphorylation (p-ERK). The positive effects of ligustilide on p-ERK, cell viability and EPO were blocked by PD98059, but not LY294002 and SB203580. In addition, transfection of SH-SY5Y cells with RTP801 plasmid increased RTP801 and LDH release, while ligustilide inhibited the effects of transfection on RTP801 expression and also increased cell viability. CONCLUSION AND IMPLICATIONS Ligustilide exerts neuroprotective effects against I/R injury by promoting EPO transcription via an ERK signalling pathway and inhibiting RTP801 expression, This compound could be developed into a therapeutic agent to prevent and treat ischaemic disorders.
Collapse
Affiliation(s)
- Xiao-mei Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | | | | | | | | | | | | |
Collapse
|
22
|
Du F, Fan M, Gong Q, Zhu LL, Zhu ZJ, Lu L, Ke Y. Effects of hypoxic preconditioning on the expression of iron influx and efflux proteins in primary neuron culture. Neurochem Int 2012; 60:335-43. [DOI: 10.1016/j.neuint.2012.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 01/05/2012] [Accepted: 01/10/2012] [Indexed: 12/01/2022]
|
23
|
Sims B, Clarke M, Francillion L, Kindred E, Hopkins ES, Sontheimer H. Hypoxic preconditioning involves system Xc- regulation in mouse neural stem cells. Stem Cell Res 2011; 8:285-91. [PMID: 22056639 DOI: 10.1016/j.scr.2011.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 08/08/2011] [Accepted: 09/19/2011] [Indexed: 10/17/2022] Open
Abstract
In animals, hypoxic preconditioning has been used as a form of neuroprotection. The exact mechanism involved in neuroprotective hypoxic preconditioning has not been described, yet could be valuable for possible neuroprotective strategies. The overexpression of the cystine-glutamate exchanger, system Xc-, has been demonstrated as being neuroprotective (Shih, Erb et al. 2006). Here, using immunohistochemistry, we demonstrate that C57BL/6 mice exposed to hypoxia showed an increase in system Xc- expression, with the highest level of intensity in the hippocampus. Western Blot analysis also showed an almost 2-fold increase in system Xc- protein in hypoxia-exposed versus control mice. The mRNA for the regulatory subunit of system Xc-, xCT, and the xCT/actin ratio were also increased under hypoxic conditions. Experiments using hypoxia-inducible factor (HIF-1α) siRNA showed a statistically significant decrease in HIF-1α and system Xc- expression. Under hypoxic conditions, system Xc- activity, as determined by cystine uptake, increased 2-fold. Importantly, hypoxic preconditioning was attenuated in neural stem cells by pharmacological inhibition of system Xc- activity with S4-carboxyphenylglycine. These data provide the first evidence of hypoxic regulation of the cystine glutamate exchanger system Xc-.
Collapse
Affiliation(s)
- Brian Sims
- Department of Pediatrics, University of Alabama at Birmingham, 35294, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Zhao ST, Huang XT, Zhang C, Ke Y. Humanin protects cortical neurons from ischemia and reperfusion injury by the increased activity of superoxide dismutase. Neurochem Res 2011; 37:153-60. [PMID: 21935731 DOI: 10.1007/s11064-011-0593-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 08/17/2011] [Accepted: 08/29/2011] [Indexed: 11/28/2022]
Abstract
The neuroprotective effects of superoxide dismutase (SOD) against hypoxia/reperfusion (I/R) injury and of humanin (HN) against toxicity by familial amyotrophic lateral sclerosis (ALS)-related mutant SOD led us to hypothesize that HN might have a role to increase the activity of SOD, which might be involved in the protective effects of HN on neuron against Alzheimer's disease-unrelated neurotoxicities. In the present study, we found that 4 h ischemia and 24 h reperfusion induced a significant increase in lactate dehydrogenase (LDH) release, malondialdehyde (MDA) formation and the number of karyopyknotic nuclei (4',6-diamidino-2-phenylindole dihydrochloride nuclear dyeing) and a decrease in the number of Calcein-AM-positive living cells and cell viability. Pretreatment of the cells with HN led to a significant decrease in LDH release, MDA formation and the number of karyopyknotic nuclei, and an increase in the number of Calcein-AM-positive living cells and cell viability in neurons treated with I/R. We also found a significant decrease in SOD activity in neurons treated with I/R only, while pre-treatment with HN before I/R induced a significant increase in the activity of SOD as compared with the I/R group. Our findings implied that HN protects cortical neurons from I/R injury by the increased SOD activity and that the protective effect of HN on neurons against I/R is concentration-dependent.
Collapse
Affiliation(s)
- Shen-Ting Zhao
- Department of Physiology, Guangzhou Medical School, Guangzhou 510182, People's Republic of China
| | | | | | | |
Collapse
|
25
|
Du F, Wu XM, Gong Q, He X, Ke Y. Hyperthermia conditioned astrocyte-cultured medium protects neurons from ischemic injury by the up-regulation of HIF-1 alpha and the increased anti-apoptotic ability. Eur J Pharmacol 2011; 666:19-25. [PMID: 21620821 DOI: 10.1016/j.ejphar.2011.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 04/20/2011] [Accepted: 05/03/2011] [Indexed: 01/08/2023]
Abstract
It has been demonstrated that conditioned medium from astrocytes challenged by in vitro ischemia (oxygen-glucose deprivation, OGD) improved neuronal survival. In addition, preconditioning stimuli can be cross-tolerant, safeguarding against other types of injury. We therefore hypothesized that hyperthermia-conditioned astrocyte-cultured medium (ACM) might also have protective effect on neurons against ischemic injury. The cultured-media, named 38ACM and 40ACM respectively, were collected after astrocytes had been incubated at 38 °C or 40 °C for 6h, followed by incubation at 37 °C for 24h. It was found that ischemia for 6h induced a significant reduction in the number of neuronal cells and cell-viability, and an increase in lactate dehydrogenase (LDH) release and the percentage of apoptotic nuclei in neurons. Pre-treatment with 38ACM or 40ACM for 24h significantly diminished ischemia injury, enhanced cell viability, reduced LDH release and reversed apoptosis. Western blot analysis showed that treatment with 38ACM or 40ACM for 24h led to a significant increase in hypoxia-inducible factor-1 (HIF-1) alpha expression. The EMSA demonstrated that the ACM increased the binding activity of HIF-1 in ischemic neurons. The data implied that hyperthermia-conditioned ACM protects neurons from ischemic injury by up-regulating HIF-1 alpha, and the increased binding activity of HIF-1 and anti-apoptotic ability.
Collapse
Affiliation(s)
- Fang Du
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | | | | | | | | |
Collapse
|
26
|
A Synergistic Role of Hyperthermic and Pharmacological Preconditioning to Protect Astrocytes Against Ischemia/Reperfusion Injury. Neurochem Res 2010; 36:312-8. [DOI: 10.1007/s11064-010-0327-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2010] [Indexed: 10/18/2022]
|
27
|
Bhuiyan MIH, Kim YJ. Mechanisms and prospects of ischemic tolerance induced by cerebral preconditioning. Int Neurourol J 2010; 14:203-12. [PMID: 21253330 PMCID: PMC3021810 DOI: 10.5213/inj.2010.14.4.203] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 12/14/2010] [Indexed: 12/20/2022] Open
Abstract
In the brain, brief episodes of ischemia induce tolerance against a subsequent severe episode of ischemia. This phenomenon of endogenous neuroprotection is known as preconditioning-induced ischemic tolerance. The purpose of this review is to summarize the current state of knowledge about mechanisms and potential applications of cerebral preconditioning and ischemic tolerance. Articles related to the terms ischemic preconditioning and ischemic tolerance were systematically searched via MEDLINE/PubMed, and articles published in English related to the nervous system were selected and analyzed. The past two decades have provided interesting insights into the molecular mechanisms of this neuroprotective phenomenon. Although both rapid and delayed types of tolerance have been documented in experimental settings, the delayed type has been found to be more prominent in the case of neuronal ischemic tolerance. Many intracellular signaling pathways have been implicated regarding ischemic preconditioning. Most of these are associated with membrane receptors, kinase cascades, and transcription factors. Moreover, ischemic tolerance can be induced by exposing animals or cells to diverse types of endogenous and exogenous stimuli that are not necessarily hypoxic or ischemic in nature. These cross-tolerances raise the hope that, in the future, it will be possible to pharmacologically activate or mimic ischemic tolerance in the human brain. Another promising approach is remote preconditioning in which preconditioning of one organ or system leads to the protection of a different (remote) organ that is difficult to target, such as the brain. The preconditioning strategy and related interventions can confer neuroprotection in experimental ischemia, and, thus, have promise for practical applications in cases of vascular neurosurgery and endo-vascular therapy.
Collapse
Affiliation(s)
| | - Youn Jung Kim
- Kyung Hee University College of Nursing Science, Seoul, Korea
| |
Collapse
|
28
|
Hyperthermic preconditioning protects astrocytes from ischemia/reperfusion injury by up-regulation of HIF-1 alpha expression and binding activity. Biochim Biophys Acta Mol Basis Dis 2010; 1802:1048-53. [DOI: 10.1016/j.bbadis.2010.06.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 06/08/2010] [Accepted: 06/18/2010] [Indexed: 12/31/2022]
|
29
|
Wu X, Qian Z, Ke Y, Du F, Zhu L. Ginkgolide B preconditioning protects neurons against ischaemia-induced apoptosis. J Cell Mol Med 2010; 13:4474-83. [PMID: 19602048 PMCID: PMC4515063 DOI: 10.1111/j.1582-4934.2008.00551.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Ischaemic preconditioning (IP) has been reported to protect the brain against subsequent lethal ischaemia, but it has not been used clinically to prevent ischaemic injury because of safety concerns. The aim of the present study was to see whether Ginkgolide B (GB) is capable of preconditioning as IP to protect neurons against ischaemic injury; if so, which mechanism is involved. Cultured mouse cortical neurons at day 8 were pre-treated with GB (120 micromol/l) for 24 hrs or exposed to short-term ischaemia (1 hr) followed by 24-hr normal culture to induce IP before being treated with severe ischaemia (5 hrs). GB and IP significantly increased cell viability, expression of hypoxia-inducible factor-1 alpha (HIF-1alpha), erythropoietin (EPO), phosphorylated Bad at serine 136 (136p-Bad) and phosphorylated glycogen synthase kinase- 3beta at serine 9 (p-GSK-3beta), and decreased the percentage of apoptotic cells and the level of active caspase-3 in severely ischaemic neurons. Moreover, LY294002 that is a specific inhibitor of phosphatidylinositol 3-kinase (PI3K) significantly reduced the enhanced expression of HIF-1alpha, EPO and 136p-Bad induced by GB and IP. These results suggest that GB, like IP in neurons, is capable of preconditioning against ischaemia-induced apoptosis, the mechanism of which may involve the PI3K signalling pathway.
Collapse
Affiliation(s)
- Xiaomei Wu
- Department of Neurochemistry, Institute for Nautical Medicine and Jiangsu Key Laboratory of Neuroregeneration, Nantong University, PRC
| | | | | | | | | |
Collapse
|
30
|
Abstract
A transient, ischemia-resistant phenotype known as "ischemic tolerance" can be established in brain in a rapid or delayed fashion by a preceding noninjurious "preconditioning" stimulus. Initial preclinical studies of this phenomenon relied primarily on brief periods of ischemia or hypoxia as preconditioning stimuli, but it was later realized that many other stressors, including pharmacologic ones, are also effective. This review highlights the surprisingly wide variety of drugs now known to promote ischemic tolerance, documented and to some extent mechanistically characterized in preclinical animal models of stroke. Although considerably more experimentation is needed to thoroughly validate the ability of any currently identified preconditioning agent to protect ischemic brain, the fact that some of these drugs are already clinically approved for other indications implies that the growing enthusiasm for translational success in the field of pharmacologic preconditioning may be well justified.
Collapse
|
31
|
Ugocsai P, Hohenstatt A, Paragh G, Liebisch G, Langmann T, Wolf Z, Weiss T, Groitl P, Dobner T, Kasprzak P, Göbölös L, Falkert A, Seelbach-Goebel B, Gellhaus A, Winterhager E, Schmidt M, Semenza GL, Schmitz G. HIF-1beta determines ABCA1 expression under hypoxia in human macrophages. Int J Biochem Cell Biol 2009; 42:241-52. [PMID: 19828131 DOI: 10.1016/j.biocel.2009.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 09/29/2009] [Accepted: 10/05/2009] [Indexed: 11/19/2022]
Abstract
ATP-binding cassette transporter A1 plays (ABCA1) a major role in reverse cholesterol transport, a process closely related to atherogenesis. In the thickening atherosclerotic lesions lipid loaded macrophages are exposed to regions of local hypoxia that may influence reverse cholesterol transport. Here we studied the effect of hypoxia on ABCA1 regulation and cholesterol efflux in human macrophages. We found that the hypoxia-inducible factor 1 (HIF-1) specifically binds to the HIF-1 response element of the ABCA1 promoter and the HIF-1 complex increases ABCA1 promoter activity along with ABCA1 expression. Primary human macrophages exposed to hypoxia or expressing constitutively active HIF-1alpha responded with a potent change in ABCA1 expression, which showed a strong correlation with HIF-1beta expression (r: 0.95-0.91). Moreover, ABCA1-mediated cholesterol efflux was also found to be regulated by HIF-1beta under hypoxia. In vivo, in macrophages prepared from human atherosclerotic lesions ABCA1 levels showed a strong correlation with HIF-1beta expression. This in vivo regulatory mechanism was confirmed in human pre-eclamptic placentas, a clinical condition with severe local hypoxia. These results demonstrate that HIF-1beta availability determines ABCA1 expression and cholesterol efflux in macrophages under hypoxia and may contribute to the interpersonal variability of atherosclerotic lesion progression.
Collapse
Affiliation(s)
- Peter Ugocsai
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Du F, Qian ZM, Zhu L, Wu XM, Yung WH, Tsim TY, Ke Y. L-DOPA neurotoxicity is mediated by up-regulation of DMT1-IRE expression. PLoS One 2009; 4:e4593. [PMID: 19240805 PMCID: PMC2643485 DOI: 10.1371/journal.pone.0004593] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Accepted: 01/15/2009] [Indexed: 11/25/2022] Open
Abstract
Background The mechanisms underlying neurotoxicity caused by L-DOPA are not yet completely known. Based on recent findings, we speculated that the increased expression of divalent metal transporter 1 without iron-response element (DMT1−IRE) induced by L-DOPA might play a critical role in the development of L-DOPA neurotoxicity. To test this hypothesis, we investigated the effects of astrocyte-conditioned medium (ACM) and siRNA DMT-IRE on L-DOPA neurotoxicity in cortical neurons. Methods and Findings We demonstrated that neurons treated with L-DOPA have a significant dose-dependent decrease in neuronal viability (MTT Assay) and increase in iron content (using a graphite furnace atomic absorption spectrophotometer), DMT1−IRE expression (Western blot analysis) and ferrous iron (55Fe(II)) uptake. Neurons incubated in ACM with or without L-DOPA had no significant differences in their morphology, Hoechst-33342 staining or viability. Also, ACM significantly inhibited the effects of L-DOPA on neuronal iron content as well as DMT1−IRE expression. In addition, we demonstrated that infection of neurons with siRNA DMT-IRE led to a significant decrease in DMT1−IRE expression as well as L-DOPA neurotoxicity. Conclusion The up-regulation of DMT1−IRE and the increase in DMT1−IRE-mediated iron influx play a key role in L-DOPA neurotoxicity in cortical neurons.
Collapse
Affiliation(s)
- Fang Du
- Department of Physiology, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Zhong-ming Qian
- Department of Applied Biology & Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong
- Department of Neurobiology and Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, People's Republic of China
- * E-mail: (Z-mQ); (KY)
| | - Li Zhu
- Department of Applied Biology & Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong
- Department of Neurobiology and Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, People's Republic of China
| | - Xiao Mei Wu
- Department of Physiology, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Wing-ho Yung
- Department of Physiology, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Ting-yuk Tsim
- Department of Physiology, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Ya Ke
- Department of Physiology, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
- * E-mail: (Z-mQ); (KY)
| |
Collapse
|
33
|
Fang D, Li Z, Zhong-ming Q, Mei WX, Ho YW, Yuan XW, Ya K. Expression of bystin in reactive astrocytes induced by ischemia/reperfusion and chemical hypoxia in vitro. Biochim Biophys Acta Mol Basis Dis 2008; 1782:658-63. [DOI: 10.1016/j.bbadis.2008.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2008] [Revised: 09/01/2008] [Accepted: 09/16/2008] [Indexed: 11/28/2022]
|