1
|
Liu X, Hong E, Xie J, Li J, Ding B, Chen Y, Xia Z, Jiang W, Lv H, Yang B, Chen Y. Txnrd2 Attenuates Early Brain Injury by Inhibition of Oxidative Stress and Endoplasmic Reticulum Stress via Trx2/Prx3 Pathway after Intracerebral Hemorrhage in Rats. Neuroscience 2024; 545:158-170. [PMID: 38513765 DOI: 10.1016/j.neuroscience.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Thioredoxin-reductase 2 (Txnrd2) belongs to the thioredoxin-reductase family of selenoproteins and is a key antioxidant enzyme in mammalian cells to regulate redox homeostasis. Here, we reported that Txnrd2 exerted a major influence in brain damage caused by Intracerebral hemorrhage (ICH) by suppressing endoplasmic reticulum (ER) stress oxidative stress and via Trx2/Prx3 pathway. Furthermore, we demonstrated that pharmacological selenium (Se) rescued the brain damage after ICH by enhancing Txnrd2 expression. Primarily, expression and localization of Txnrd2, Trx2 and Prx3 were determined in collagenase IV-induced ICH model. Txnrd2 was then knocked down using siRNA interference in rats which were found to develop more severe encephaledema and neurological deficits. Mechanistically, we observed that loss of Txnrd2 leads to increased lipid peroxidation levels and ER stress protein expression in neurons and astrocytes. Additionally, it was revealed that Se effectively restored the expression of Txnrd2 in brain and inhibited both the activity of ER stress protein activity and the generation of reactive oxygen species (ROS) by promoting Trx2/Prx3 kilter when administrating sodium selenite in lateral ventricle. This study shed light on the effect of Txnrd2 in regulating oxidative stress and ER stress via Trx2/Prx3 pathway upon ICH and its promising potential as an ICH therapeutic target.
Collapse
Affiliation(s)
- Xuanbei Liu
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, China
| | - Enhui Hong
- Department of Neurosurgery, Jiu Jiang No.1 People's Hospital, Jiu Jiang, China
| | - Jiayu Xie
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, China
| | - Jiangwei Li
- Department of Neurosurgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Boyun Ding
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, China
| | - Yongsheng Chen
- Department of Neurosurgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhennan Xia
- Department of Neurosurgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Weiping Jiang
- Department of Neurosurgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Hongzhu Lv
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, China
| | - Bo Yang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, China
| | - Yizhao Chen
- Department of Neurosurgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Ismael S, Nasoohi S, Li L, Aslam KS, Khan MM, El-Remessy AB, McDonald MP, Liao FF, Ishrat T. Thioredoxin interacting protein regulates age-associated neuroinflammation. Neurobiol Dis 2021; 156:105399. [PMID: 34029695 PMCID: PMC8277763 DOI: 10.1016/j.nbd.2021.105399] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/07/2021] [Accepted: 05/16/2021] [Indexed: 12/15/2022] Open
Abstract
Immune system hypersensitivity is believed to contribute to mental frailty in the elderly. Solid evidence indicates NOD-like receptor pyrin domain containing-3 (NLRP3)-inflammasome activation intimately connects aging-associated chronic inflammation (inflammaging) to senile cognitive decline. Thioredoxin interacting protein (TXNIP), an inducible protein involved in oxidative stress, is essential for NLRP3 inflammasome activity. This study aims to find whether TXNIP/NLRP3 inflammasome pathway is involved in senile dementia. According to our studies on sex-matched mice, TXNIP was significantly upregulated in aged animals, paralleled by the NLRP3-inflammasome over-activity leading to enhanced caspase-1 cleavage and IL-1β maturation, in both sexes. This was closely associated with depletion of the anti-aging and cognition enhancing protein klotho, in aged males. Txnip knockout reversed age-related NLRP3-hyperactivity and enhanced thioredoxin (TRX) levels. Further, TXNIP inhibition along with verapamil replicated TXNIP/NLRP3-inflammasome downregulation in aged animals, with FOXO-1 and mTOR upregulation. These alterations concurred with substantial improvements in both cognitive and sensorimotor abilities. Together, these findings substantiate the pivotal role of TXNIP to drive inflammaging in parallel with klotho depletion and functional decline, and delineate thioredoxin system as a potential target to decelerate senile dementia.
Collapse
Affiliation(s)
- Saifudeen Ismael
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Sanaz Nasoohi
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Lexiao Li
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA; Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States of America.
| | - Khurram S Aslam
- Center for Earthquake Research and Information, University of Memphis, Memphis, TN, United States of America
| | - Mohammad Moshahid Khan
- Department of Neurology, The University of Tennessee Health Science Center, Memphis, TN, United States of America; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Azza B El-Remessy
- Department of Pharmacy, Doctors Hospital of Augusta, GA, United States of America.
| | - Michael P McDonald
- Department of Neurology, The University of Tennessee Health Science Center, Memphis, TN, United States of America; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Francesca-Fang Liao
- Department of Pharmacology, The University of Tennessee Health Science Center, Memphis, TN, United States of America; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA; Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States of America; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
3
|
Yoo YH, Kim DW, Chen BH, Sim H, Kim B, Lee JC, Ahn JH, Park Y, Cho JH, Kang IJ, Won MH, Lee TK. Comparison of age-dependent alterations in thioredoxin 2 and thioredoxin reductase 2 expressions in hippocampi between mice and rats. Lab Anim Res 2021; 37:11. [PMID: 33676586 PMCID: PMC7937215 DOI: 10.1186/s42826-021-00088-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/25/2021] [Indexed: 11/10/2022] Open
Abstract
Background Aging is one of major causes triggering neurophysiological changes in many brain substructures, including the hippocampus, which has a major role in learning and memory. Thioredoxin (Trx) is a class of small redox proteins. Among the Trx family, Trx2 plays an important role in the regulation of mitochondrial membrane potential and is controlled by TrxR2. Hitherto, age-dependent alterations in Trx2 and TrxR2 in aged hippocampi have been poorly investigated. Therefore, the aim of this study was to examine changes in Trx2 and TrxR2 in mouse and rat hippocampi by age and to compare their differences between mice and rats. Results Trx2 and TrxR2 levels using Western blots in mice were the highest at young age and gradually reduced with time, showing that no significant differences in the levels were found between the two subfields. In rats, however, their expression levels were the lowest at young age and gradually increased with time. Nevertheless, there were no differences in cellular distribution and morphology in their hippocampi when it was observed by cresyl violet staining. In addition, both Trx2 and TrxR2 immunoreactivities in the CA1-3 fields were mainly shown in pyramidal cells (principal cells), showing that their immunoreactivities were altered like changes in their protein levels. Conclusions Our current findings suggest that Trx2 and TrxR2 expressions in the brain may be different according to brain regions, age and species. Therefore, further studies are needed to examine the reasons of the differences of Trx2 and TrxR2 expressions in the hippocampus between mice and rats.
Collapse
Affiliation(s)
- Yeon Ho Yoo
- Department of Emergency Medicine, Institute of Medical Sciences, School of Medicine, Kangwon National University Hospital, Kangwon National University, 24289, Chuncheon, Gangwon, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, 25457, Gangneung, Gangwon, Republic of Korea
| | - Bai Hui Chen
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, P.R. China
| | - Hyejin Sim
- Department of Neurobiology, School of Medicine, Kangwon National University, 24341, Chuncheon, Gangwon, Republic of Korea
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, 24341, Chuncheon, Gangwon, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, 24341, Chuncheon, Gangwon, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, 24341, Chuncheon, Gangwon, Republic of Korea.,Department of Physical Therapy, College of Health Science, Youngsan University, 50510, Yangsan, Gyeongnam, Republic of Korea
| | - Yoonsoo Park
- Department of Emergency Medicine, Institute of Medical Sciences, School of Medicine, Kangwon National University Hospital, Kangwon National University, 24289, Chuncheon, Gangwon, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, Institute of Medical Sciences, School of Medicine, Kangwon National University Hospital, Kangwon National University, 24289, Chuncheon, Gangwon, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, 24252, Chuncheon, Gangwon, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, 24341, Chuncheon, Gangwon, Republic of Korea.
| | - Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 24252, Chuncheon, Gangwon, Republic of Korea.
| |
Collapse
|
4
|
Kiffer F, Alexander T, Anderson J, Groves T, McElroy T, Wang J, Sridharan V, Bauer M, Boerma M, Allen A. Late Effects of 1H + 16O on Short-Term and Object Memory, Hippocampal Dendritic Morphology and Mutagenesis. Front Behav Neurosci 2020; 14:96. [PMID: 32670032 PMCID: PMC7332779 DOI: 10.3389/fnbeh.2020.00096] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/22/2020] [Indexed: 11/17/2022] Open
Abstract
The space extending beyond Earth’s magnetosphere is subject to a complex field of high-energy charged nuclei, which are capable of traversing spacecraft shielding and human tissues, inducing dense ionization events. The central nervous system is a major area of concern for astronauts who will be exposed to the deep-space radiation environment on a mission to Mars, as charged-particle radiation has been shown to elicit changes to the dendritic arbor within the hippocampus of rodents, and related cognitive-behavioral deficits. We exposed 6-month-old male mice to whole-body 1H (0.5 Gy; 150 MeV/n; 18–19 cGy/minute) and an hour later to 16O (0.1Gy; 600 MeV/n; 18–33 Gy/min) at NASA’s Space Radiation Laboratory as a galactic cosmic ray-relevant model. Animals were housed with bedding which provides cognitive enrichment. Mice were tested for cognitive behavior 9 months after exposure to elucidate late radiation effects. Radiation induced significant deficits in novel object recognition and short-term spatial memory (Y-maze). Additionally, we observed opposing morphological differences between the mature granular and pyramidal neurons throughout the hippocampus, with increased dendritic length in the dorsal dentate gyrus and reduced length and complexity in the CA1 subregion of the hippocampus. Dendritic spine analyses revealed a severe reduction in mushroom spine density throughout the hippocampus of irradiated animals. Finally, we detected no general effect of radiation on single-nucleotide polymorphisms in immediate early genes, and genes involved in inflammation but found a higher variant allele frequency in the antioxidants thioredoxin reductase 2 and 3 loci.
Collapse
Affiliation(s)
- Frederico Kiffer
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Tyler Alexander
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Julie Anderson
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Thomas Groves
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Michael Bauer
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antiño Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
5
|
Qin Y, Zhang Q, Liu Y. Analysis of knowledge bases and research focuses of cerebral ischemia-reperfusion from the perspective of mapping knowledge domain. Brain Res Bull 2019; 156:15-24. [PMID: 31843561 DOI: 10.1016/j.brainresbull.2019.12.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 11/17/2022]
Abstract
Cerebral ischemia-reperfusion (IR) has attracted wide attention as a serious clinical problem. So far, the field has accumulated a large amount of scientific research literature. To clarify the temporal and spatial distribution characteristics of research resources, knowledge bases and research focuses, a visual analysis was performed on 5814 articles cited in the WoS databases from 2004 to 2019. This analysis was based on bibliometrics and mapping knowledge domain (MKD) analysis with VOSviewer, and CiteSpace 5.4.R4. The results can be elaborated from four aspects. First, the volume of publications in this area is on the rise. Second, the United States and China are the active regions. The USA is the central region of cerebral ischemia-reperfusion research. Third, the knowledge bases of IR have focused on five major areas of "Suitable small-animal models", "A framework with further study", "Molecular signaling targets by oxidative stress", "Finding new potential targets for therapy" and "Protective effect of multiple transient ischemia". Fourth, the research focuses consist of three representative areas: "Oxidative stress closelyd with cerebral ischemia-reperfusion", "Neuronal apoptosis and neuronal protection", and "Neuroprotective effect of the blood-brain barrier".
Collapse
Affiliation(s)
- Yi Qin
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China; Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Qing Zhang
- No.4 Hospital Beijing University of Chinese Medicine, Zaozhuang, Shandong 277000
| | - Yaru Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
6
|
Duan Q, Si E. MicroRNA-25 aggravates Aβ1-42-induced hippocampal neuron injury in Alzheimer's disease by downregulating KLF2 via the Nrf2 signaling pathway in a mouse model. J Cell Biochem 2019; 120:15891-15905. [PMID: 31144355 DOI: 10.1002/jcb.28861] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/21/2022]
Abstract
Recently, numerous microRNAs (miRNAs) have been considered as key players in the regulation of neuronal processes. The purpose of the present study is to explore the effect of miR-25 on hippocampal neuron injury in Alzheimer's disease (AD) induced by amyloid β (Aβ) peptide fragment 1 to 42 (Aβ1-42) via Kruppel-like factor 2 (KLF2) through the nuclear factor-E2-related factor 2 (Nrf2) signaling pathway. A mouse model of AD was established through Aβ1-42 induction. The underlying regulatory mechanisms of miR-25 were analyzed through treatment of miR-25 mimics, miR-25 inhibitors, or small interfering RNA (siRNA) against KLF2 in hippocampal tissues and cells isolated from AD mice. The targeting relationship between miR-25 and KLF2 was predicted using a target prediction program and verified by luciferase activity determination. MTT assay was used to evaluate the proliferative ability and flow cytometry to detect cell cycle distribution and apoptosis. KLF2 was confirmed as a target gene of miR-25. When the mice were induced by Aβ1-42, proliferation was suppressed while apoptosis was promoted in hippocampal neurons as evidenced by lower levels of KLF2, Nrf2, haem oxygenase, glutathione S transferase α1, glutathione, thioredoxin, and B-cell lymphoma-2 along with higher bax level. However, such alternations could be reversed by treatment of miR-25 inhibitors. These findings indicate that miR-25 may inhibit hippocampal neuron proliferation while promoting apoptosis, thereby aggravating hippocampal neuron injury through downregulation of KLF2 via the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Qiang Duan
- Department of Neurology, Heze Third People's Hospital, Heze, People's Republic of China
| | - Erwang Si
- Department of Neurology, Heze Third People's Hospital, Heze, People's Republic of China
| |
Collapse
|
7
|
Fan DX, Yang XH, Li YN, Guo L. 17β-Estradiol on the Expression of G-Protein Coupled Estrogen Receptor (GPER/GPR30) Mitophagy, and the PI3K/Akt Signaling Pathway in ATDC5 Chondrocytes In Vitro. Med Sci Monit 2018; 24:1936-1947. [PMID: 29608013 PMCID: PMC5898603 DOI: 10.12659/msm.909365] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Osteoarthritis is a progressive inflammatory joint disease resulting in damage to articular cartilage. G-protein coupled estrogen receptor (GPER/GPR30) activates cell signaling in response to 17β-estradiol, which can be blocked by the GPR30 agonist, G15, an analog of G-1. The aims of this study were to investigate the effects of 17β-estradiol on the expression of G-protein coupled estrogen receptor (GPER/GPR30) on mitophagy and the PI3K/Akt signaling pathway in ATDC5 chondrocytes in vitro. Material/Methods Cultured ATDC5 chondrocytes were treated with increasing concentrations of 17β-estradiol with and without G15, p38 inhibitor (SB203580), JNK inhibitor (SP600125), PI3K inhibitor (LY294002, S1737), and mTOR inhibitor (S1842). Expression of GPER/GPR30 and components of the PI3K/Akt pathway in cultured ATDC5 chondrocytes were detected by immunofluorescence (IF) staining, Western blot, and real-time polymerase chain reaction (RT-PCR). Transmission electron microscopy (TEM) and IF were used to detect mitophagosomes. Expression of LC-3, LAMP2, TOM20, Hsp60, p-Akt, p-mTOR, p-p38, and p-JNK was investigated by Western blot. Proliferation and viability of the ATDC5 chondrocytes were determined using BrdU and MTT assays. Results In 17β-estradiol-treated ATDC5 chondrocytes, increased expression of GPER/GPR30 was found, but fewer mitophagosomes were observed, and decreased numbers of TOM20-positive granules were co-localized with decreased LAMP2 and increased expression levels of TOM20, Hsp60, p-Akt, and p-mTOR, and reduced expression of LC3-II, were found. In 17β-estradiol-treated ATDC5 chondrocytes, the proliferation and viability of the 17β-estradiol-treated ATDC5 chondrocytes were significantly elevated. Conclusions Treatment with 17β-estradiol protected ATDC5 chondrocytes against mitophagy via the GPER/GPR30 and the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Dong-Xiao Fan
- Department of Orthopedic Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China (mainland).,Orthopedic Surgery, First Affiliated Hospital, China Medical University, , China (mainland)
| | - Xu-Hao Yang
- Department of Orthopedic Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yi-Nan Li
- Department of Orthopedic Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| | - Lei Guo
- Department of Orthopedic Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
8
|
Wang Z, Ye Z, Huang G, Wang N, Wang E, Guo Q. Sevoflurane Post-conditioning Enhanced Hippocampal Neuron Resistance to Global Cerebral Ischemia Induced by Cardiac Arrest in Rats through PI3K/Akt Survival Pathway. Front Cell Neurosci 2016; 10:271. [PMID: 27965539 PMCID: PMC5127837 DOI: 10.3389/fncel.2016.00271] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022] Open
Abstract
The purpose of this current study was to evaluate whether improvement of mitochondrial dysfunction was involved in the therapeutic effect of sevoflurane post-conditioning in global cerebral ischemia after cardiac arrest (CA) via the PI3K/Akt pathway. In the first experiment, animals were randomly divided into three groups: a sham group, a CA group, a CA+sevoflurane post-conditioning group (CA+SE). Sevoflurane post-conditioning was achieved by administration of 2.5% sevoflurane for 30 min after resuscitation. Sevoflurane post-conditioning has a significant neuroprotective effect by increasing survival rates and reducing neuronal apoptosis. Additionally, the gene and protein expression of PGC-1α, NRF-1, and TFAM, the master regulators of mitochondrial biogenesis, were up-regulated in the CA+SE group, when compared to the CA group. Similarly, in contrast to the CA group, mitochondria-specific antioxidant enzymes, including heat-shock protein 60 (HSP60), peroxiredoxin 3 (Prx3), and thioredoxin 2 (Trx2) were also increased in the CA+SE group. Finally, administration of sevoflurane ameliorated mitochondrial reactive oxygen species (ROS) formation and maintained mitochondrial integrity. In the second experiment, we investigated the relationship between the PI3K/Akt pathway and mitochondrial biogenesis and mitochondria-specific antioxidant enzymes in sevoflurane-induced neuroprotection. The selective PI3K inhibitor wortmannin not only eliminated the beneficial biochemical processes of sevoflurane by reducing the level of mitochondrial biogenesis-related proteins and aggravating mitochondrial integrity, but also reversed the elevation of mitochondria-specific antioxidant enzymes induced by sevoflurane. Therefore, our data suggested that sevoflurane post-conditioning provides neuroprotection via improving mitochondrial biogenesis and integrity, as well as increasing mitochondria-specific antioxidant enzymes by a mechanism involving the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South UniversityChangsha, China; Department of Anesthesiology, Hainan General HospitalHaikou, China
| | - Zhi Ye
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University Changsha, China
| | - Guoqing Huang
- Emergency Department, Affiliated Xiangya Hospital of Central South University Changsha, China
| | - Na Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University Changsha, China
| | - E Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University Changsha, China
| |
Collapse
|