1
|
Yao H, Tian J, Cheng S, Dou H, Zhu Y. The mechanism of hypoxia-inducible factor-1α enhancing the transcriptional activity of transferrin ferroportin 1 and regulating the Nrf2/HO-1 pathway in ferroptosis after cerebral ischemic injury. Neuroscience 2024; 559:26-38. [PMID: 39168172 DOI: 10.1016/j.neuroscience.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 07/05/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Cerebral ischemic/reperfusion (I/R) injury has high disability and morbidity. Hypoxia-inducible factor-1α (HIF-1α) may enhance the transcriptional activity of transferrin ferroportin 1 (FPN1) in regulating ferroptosis after cerebral ischemia injury (CII). In this study, cerebral I/R injury rat models were established and treated with pcDNA3.1-HIF-1α, pcDNA3.1-NC lentiviral plasmid, or ML385 (a specific Nrf2 inhibitor). Additionally, oxygen-glucose deprivation/reoxygenation (OGD/R) exposed PC12 cells were used as an in vitro model of cerebral ischemia and treated with pcDNA3.1-HIF-1α, si-FPN1, or ML385. The results elicited that cerebral I/R injury rats exhibited increased Longa scores, TUNEL and NeuN co-positive cells, Fe2+ concentration, ROS and HIF-1α levels, and MDA content, while reduced cell density and number, GSH content, and GPX4 protein level. Morphologically abnormal and disordered hippocampal neurons were also observed in CII rats. HIF-1α inhibited brain neuron ferroptosis and ameliorated I/R injury. HIF-1α alleviated OGD-induced PC12 cell ferroptosis. OGD/R decreased FPN1 protein level in PC12 cells, and HIF-1α enhanced FPN1 transcriptional activity. FPN1 knockdown reversed HIF-1α-mediated alleviation of OGD/R-induced ferroptosis. HIF-1α activated the Nrf2/HO-1 pathway by enhancing FPN1 expression and alleviating OGD/R-induced ferroptosis. Conjointly, HIF-1α enhanced the transcriptional activity of FPN1, activated the Nrf2/HO-1 pathway, and inhibited ferroptosis of brain neurons, thereby improving I/R injury in CII rats.
Collapse
Affiliation(s)
- Haiqian Yao
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Jianan Tian
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Shi Cheng
- Department of Orthopaedics, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Haitong Dou
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Yulan Zhu
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
2
|
Chen C, Song C, Liu B, Wang Y, Jia J, Pang K, Wang Y, Wang P. Activation of BMP4/SMAD pathway by HIF-1α in hypoxic environment promotes osteogenic differentiation of BMSCs and leads to ectopic bone formation. Tissue Cell 2024; 88:102376. [PMID: 38608407 DOI: 10.1016/j.tice.2024.102376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 03/15/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVE Heterotopic ossification (HO), also known as ossifying myositis, is a condition that produces abnormal bone and cartilage tissue in the soft tissues. Hypoxia inducible factor lα (HIF-lα) regulates the expression of various genes, which is closely related to the promotion of bone formation, and Drosophila mothers against decapentaplegic protein (SMAD) mediates the signal transduction in the Bone morphogenetic protein (BMP) signaling pathway, which affects the function of osteoblasts and osteoclasts, and thus plays a key role in the regulation of bone remodeling. We aimed to investigate the mechanism by which HIF-1α induces osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in a hypoxic environment. METHODS A cellular hypoxia model was constructed to verify the expression of HIF-1α, while alizarin red staining was performed to observe the osteogenic differentiation ability of bone marrow mesenchymal stem cells (BMSCs). Alizarin red staining was used to analyze the late mineralization ability of the cells. Western blot analysis was performed to analyze the expression levels of osteogenesis-related factors OCN, OPN proteins as well as the pathway proteins BMP4, p-Smad1/5/8, and Smad1. We also constructed a rat model of ectopic bone formation, observed ectopic ossification by X-ray, and verified the success of the rat model by ELISA of HIF-1α. HE staining was used to observe the matrix and trabecular structure of bone, and Masson staining was used to observe the collagen and trabecular structure of bone. Immunohistochemistry analyzed the expression of OCN and OPN in ectopic bone tissues, and WB analyzed the expression of pathway proteins BMP4, p-Smad1/5/8 and Smad1 in ectopic bone tissues to verify the signaling pathway of ectopic bone formation. RESULTS Our results indicate that hypoxic environment upregulates HIF-1a expression and activates BMP4/SMAD signaling pathway. This led to an increase in ALP content and enhanced expression of the osteogenesis-related factors OCN and OPN, resulting in enhanced osteogenic differentiation of BMSCs. The results of our in vivo experiments showed that rats inoculated with BMSCs overexpressing HIF-1α showed bony structures in tendon tissues, enhanced expression of the bone signaling pathways BMP4 and p-Smad1/5/8, and enhanced expression levels of the osteogenic-related factors OCN and OPN, resulting in the formation of ectopic bone. CONCLUSIONS These data further suggest a novel mechanistic view that hypoxic bone marrow BMSCs activate the BMP4/SMAD pathway by up-regulating the expression level of HIF-1α, thereby promoting the secretion of osteogenic factors leading to ectopic bone formation.
Collapse
Affiliation(s)
- Cong Chen
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Chunhao Song
- Department of Medical Imaging, Weihai Wendeng District People Hospital, Weihai 264200, China
| | - Bo Liu
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Yitao Wang
- Department of Laboratory, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Jun Jia
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Kai Pang
- Department of Operations Management, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Yuanhao Wang
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Peng Wang
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China.
| |
Collapse
|
3
|
Ma H, Ye D, Liu Y, Wu P, Yu L, Guo L, Gao Y, Liu Y, Yan H, Shi J. Propofol suppresses OGD/R-induced ferroptosis in neurons by inhibiting the HIF-1α/YTHDF1/BECN1 axis. Brain Inj 2023; 37:1285-1293. [PMID: 37614036 DOI: 10.1080/02699052.2023.2237881] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 06/07/2023] [Accepted: 07/03/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Ischemia/reperfusion (I/R) is a pathological process that causes severe damage. Propofol is known to alleviate I/R-related injury; however, the exact function and underlying mechanisms are not fully understood. METHODS Using an oxygen glucose deprivation/re-oxygenation (OGD/R) method, an in vitro I/R injury model was induced. The cell viability and the level of Fe2+, glutathione synthetase (GSH), and malondialdehyde (MDA) were evaluated using kits. Luciferase reporter gene assay, chromatin immunoprecipitation, and RNA immunoprecipitation (RIP) were used to verify the interaction between molecules. The m6A level of BECN1 mRNA was determined through methylated RIP. RESULTS Propofol-treated OGD/R models showed reduced levels of Fe2+ and MDA, while the cell viability and the level of GSH increased. Propofol inhibited ferroptosis by down-regulating HIF-1α in OGD/R-treated HT22 cells. HIF-1α is bound to the promoter region of YTHDF1 to promote its transcription, and YTHDF1 promoted ferroptosis by stabilizing the mRNA of BECN1. The suppressive effect of propofol on OGD/R-induced ferroptosis was reversed by the overexpression of YTHDF1. CONCLUSIONS Our study revealed that the HIF-1α/YTHDF1/BECN1 axis in OGD/R-treated HT22 cells promotes ferroptosis, and administration of propofol can inhibit this axis to avoid cell death. This study provides a novel insight for the neuroprotective function of propofol.
Collapse
Affiliation(s)
- Hongyan Ma
- Department of Anesthesiology (Qunli Campus), The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Dongxue Ye
- Department of Anesthesiology (Qunli Campus), The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Yuqing Liu
- Department of Anesthesiology (Qunli Campus), The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Pei Wu
- Department of Neurosrugery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Lu Yu
- Department of Anesthesiology (Qunli Campus), The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Libo Guo
- Department of Anesthesiology (Qunli Campus), The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Yang Gao
- Department of Anesthesiology (Qunli Campus), The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Ying Liu
- Department of Anesthesiology (Qunli Campus), The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Haiyan Yan
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| | - Jinghui Shi
- Department of Anesthesiology (Qunli Campus), The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, P.R China
| |
Collapse
|
4
|
Neag MA, Mitre AO, Burlacu CC, Inceu AI, Mihu C, Melincovici CS, Bichescu M, Buzoianu AD. miRNA Involvement in Cerebral Ischemia-Reperfusion Injury. Front Neurosci 2022; 16:901360. [PMID: 35757539 PMCID: PMC9226476 DOI: 10.3389/fnins.2022.901360] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral ischemia reperfusion injury is a debilitating medical condition, currently with only a limited amount of therapies aimed at protecting the cerebral parenchyma. Micro RNAs (miRNAs) are small, non-coding RNA molecules that via the RNA-induced silencing complex either degrade or prevent target messenger RNAs from being translated and thus, can modulate the synthesis of target proteins. In the neurological field, miRNAs have been evaluated as potential regulators in brain development processes and pathological events. Following ischemic hypoxic stress, the cellular and molecular events initiated dysregulate different miRNAs, responsible for long-terming progression and extension of neuronal damage. Because of their ability to regulate the synthesis of target proteins, miRNAs emerge as a possible therapeutic strategy in limiting the neuronal damage following a cerebral ischemic event. This review aims to summarize the recent literature evidence of the miRNAs involved in signaling and modulating cerebral ischemia-reperfusion injuries, thus pointing their potential in limiting neuronal damage and repair mechanisms. An in-depth overview of the molecular pathways involved in ischemia reperfusion injury and the involvement of specific miRNAs, could provide future perspectives in the development of neuroprotective agents targeting these specific miRNAs.
Collapse
Affiliation(s)
- Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andrei-Otto Mitre
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Andreea-Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Carina Mihu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Carmen-Stanca Melincovici
- Department of Morphological Sciences, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Marius Bichescu
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
5
|
Yang J, Xie X. Tofacitinib protects intestinal epithelial cells against oxygen-glucose deprivation/reoxygenation injury by inhibiting the JAK/STAT3 signaling pathway. Exp Ther Med 2021; 22:1108. [PMID: 34504562 PMCID: PMC8383770 DOI: 10.3892/etm.2021.10542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the role and potential mechanism of action of tofacitinib (Tofa) in intestinal ischemia/reperfusion (I/R) injury. The normal rat small intestine epithelial cell line, IEC-6, was used to establish an I/R injury model by inducing oxygen-glucose deprivation/reoxygenation (OGD/R). Cells were divided into the following five groups: Control, OGD/R, OGD/R with 50, 100 and 200 nM Tofa. Following Tofa administration, cell viability was measured using Cell Counting Kit-8 assay and a lactate dehydrogenase detection kit. The expression levels of cell apoptosis-related proteins, Bcl-2, cleaved-caspase-3 and cleaved-caspase-9 were detected using western blot analysis. Additionally, the levels of oxidative stress-related markers, such as reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD), and inflammatory cytokines, TNF-α, IL-6 and IL-1β were assessed using the colorimetric method. Western blot analysis was also used to measure the expression levels of the Janus kinase (JAK)/STAT3 signaling pathway-related proteins, including phosphorylated (p)-JAK1, p-JAK3 and p-STAT3. Subsequently, colivelin, an agonist of the JAK/STAT3 pathway, was used to investigate whether the effects of Tofa on intestinal I/R injury were mediated by this signaling pathway. The results showed that Tofa dose-dependently elevated cell viability compared with that in the OGD/R group. By contrast, Tofa attenuated cell apoptosis, which was coupled with upregulated Bcl-2 expression, downregulated cleaved-caspase-3 and downregulated cleaved-caspase-9 levels, in OGD/R-induced IEC-6 cells. Furthermore, the contents of ROS and MDA were significantly increased following exposure to OGD/R, which were accompanied by the decreased activity of SOD. These effects were reversed following cell treatment with Tofa. Consistently, Tofa intervention reduced the secretion levels of TNF-α, IL-6 and IL-1β in a dose-dependent manner. Additionally, Tofa markedly downregulated the phosphorylation levels of JAK1, JAK3 and STAT3 in OGD/R-induced IEC-6 cells. However, treatment with colivelin markedly reversed the inhibitory effects of Tofa on cell viability, cell apoptosis, oxidative stress and inflammation. Overall, the findings of the present study suggested that Tofa could protect against intestinal I/R injury by inhibiting the JAK/STAT3 signaling pathway, which may hold promise as a therapeutic agent for intestinal I/R injury.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pediatric Gastroenterology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610091, P.R. China
| | - Xiaoli Xie
- Department of Pediatric Gastroenterology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610091, P.R. China
| |
Collapse
|
6
|
Fang H, Li HF, He MH, Yang M, Zhang JP. HDAC3 Downregulation Improves Cerebral Ischemic Injury via Regulation of the SDC1-Dependent JAK1/STAT3 Signaling Pathway Through miR-19a Upregulation. Mol Neurobiol 2021; 58:3158-3174. [PMID: 33634377 DOI: 10.1007/s12035-021-02325-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 02/08/2021] [Indexed: 12/28/2022]
Abstract
Histone deacetylase (HDAC) inhibitors can protect the brain from ischemic injury. This study aimed to identify the regulation of HDAC3 in cerebral ischemic injury. Middle cerebral artery occlusion (MCAO) was performed to establish a mouse model with cerebral ischemic injury, in which expression of HDAC3 and miR-19a was evaluated using RT-qPCR. In MCAO mice with silencing of HDAC3, infarct volume was determined using 2,3,5-triphenyl tetrazolium chloride (TTC) staining, and serum levels of TNF-α, IL-6, and IL-8 were measured using ELISA. An in vitro model was constructed in human umbilical vein endothelial cells (HUVECs) with oxygen-glucose deprivation/reoxygenation (OGD/R), followed by gain- and loss-of-function experiments. Relationships among miR-19a, HDAC3, and syndecan-1 (SDC1) were explored using RIP, ChIP, and dual-luciferase reporter assays. The expression of HDAC3, SDC1, JAK1, and STAT3 along with the extent of JAK1 and STAT3 phosphorylation was measured by Western blot analysis. HUVEC viability, apoptosis, and angiogenesis were assessed by CCK-8, flow cytometry, and angiogenesis assays in vitro separately. We found elevated HDAC3 and downregulated miR-19a expression in the MCAO mice. Decreased TNF-α, IL-6, and IL-8 serum levels were observed in response to silencing of HDAC3. HDAC3 inhibited the expression of miR-19a, which in turn targeted SDC1, leading to JAK1/STAT3 signaling pathway activation. HDAC3 overexpression or miR-19a inhibition repressed HUVEC viability and angiogenesis but enhanced HUVEC apoptosis. Our data unraveled the mechanism whereby HDAC3 inhibition ameliorated cerebral ischemic injury by activating the JAK1/STAT3 signaling pathway through miR-19a-mediated SDC1 inhibition.
Collapse
Affiliation(s)
- Hua Fang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, No. 83, Zhongshan East Road, Guiyang, 550002, People's Republic of China
- Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, 550002, People's Republic of China
- Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, 550025, People's Republic of China
| | - Hua-Feng Li
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Ming-Hai He
- Department of Anesthesiology, Guizhou Provincial People's Hospital, No. 83, Zhongshan East Road, Guiyang, 550002, People's Republic of China
- Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, 550002, People's Republic of China
- Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, 550025, People's Republic of China
| | - Miao Yang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, No. 83, Zhongshan East Road, Guiyang, 550002, People's Republic of China
- Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, 550002, People's Republic of China
- Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, 550025, People's Republic of China
| | - Jian-Ping Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, No. 83, Zhongshan East Road, Guiyang, 550002, People's Republic of China.
- Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, 550002, People's Republic of China.
- Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, 550025, People's Republic of China.
| |
Collapse
|
7
|
Li YJ, Zhang DZ, Xi Y, Wu CA. Protective effect of dexmedetomidine on neuronal hypoxic injury through inhibition of miR-134. Hum Exp Toxicol 2021; 40:2145-2155. [PMID: 34121490 DOI: 10.1177/09603271211023784] [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] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To explore the mechanism of dexmedetomidine (DEX)-mediated miR-134 inhibition in hypoxia-induced damage in PC12 cells. METHODS Hydrogen peroxide (H2O2)-stimulated PC12 cells were divided into control, H2O2, DEX + H2O2, miR-NC/inhibitor + H2O2, and miR-NC/ mimic + DEX + H2O2 groups. Cell viability and apoptosis were assessed by the 3-(4,5-dimethylthiazol(-2-y1)-2,5-diphenytetrazolium bromide (MTT) assay and Annexin V-FITC/PI staining, while gene and protein expression levels were detected by qRT-PCR and western blotting. Reactive oxygen species (ROS) levels were tested by 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) staining, and malondialdehyde (MDA) content was determined with a detection kit. RESULTS DEX treatment decreased H2O2-elevated miR-134 expression. H2O2-induced PC12 cell damage was improved by DEX and miR-134 inhibitor; additionally, cell viability was increased, while cell apoptosis was reduced. In addition, both DEX and miR-134 inhibitor reduced the upregulated expression of cleaved caspase-3 and increased the downregulated expression of Bcl-2 in H2O2-induced PC12 cells. However, compared to that in the DEX + H2O2 group, cell viability in the mimic + DEX + H2O2 group was decreased, and the apoptotic rate was elevated with increased cleaved caspase-3 and decreased Bcl-2 expression. Inflammation and oxidative stress were increased in H2O2-induced PC12 cells but improved with DEX or miR-134 inhibitor treatment. However, this improvement of H2O2-induced inflammation and oxidative stress induced by DEX in PC12 cells could be reversed by the miR-134 mimic. CONCLUSION DEX exerts protective effects to promote viability and reduce cell apoptosis, inflammation, and oxidative stress in H2O2-induced PC12 cells by inhibiting the expression of miR-134.
Collapse
Affiliation(s)
- Y-J Li
- Department of Anesthesiology, Beijing Jishuitan Hospital, Beijing, China
| | - D-Z Zhang
- Department of Anesthesiology, Beijing Jishuitan Hospital, Beijing, China
| | - Y Xi
- Department of Anesthesiology, Beijing Jishuitan Hospital, Beijing, China
| | - C-A Wu
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| |
Collapse
|
8
|
Dendrobium nobile Lindl. polysaccharides reduce cerebral ischemia/reperfusion injury in mice by increasing myeloid cell leukemia 1 via the downregulation of miR-134. Neuroreport 2020; 32:177-187. [PMID: 33323840 DOI: 10.1097/wnr.0000000000001562] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE The traditional Chinese medicine, Dendrobium nobile Lindl. polysaccharides (DNLP), reportedly has neuroprotective effects. However, its effects following ischemic stroke remain unclear. This study aimed to explore the role and mechanism of DNLP in experimental models of inflammation and apoptosis. METHODS Inflammation and apoptosis were induced by in vivo ischemia/reperfusion and by in vitro oxygen glucose deprivation/reperfusion (OGD/R). In the in vivo model, immediately after the induction of cerebral ischemia, 50, 100, and 200 mg/kg DNLP were injected intraperitoneally. We subsequently detected indicators of neuronal damage. RESULTS Treatment with Dendrobium nobile Lindl. polysaccharide significantly reduced cerebral ischemic injury. After in vivo and in vitro middle cerebral artery occlusion/reperfusion or OGD/R-induced hypoxia injury, miR-134 expression in neurons was significantly increased. Altering the expression of miR-134 induced changes in myeloid cell leukemia 1 (MCL-1), one of its target proteins. In addition, DNLP significantly downregulated the in vivo and in vitro expression of miR-134 after ischemic injury, and influenced inflammation and apoptotic proteins by altering the level of MCL-1 protein. DNLP also had a protective effect on neurons damaged by OGD/R, which could improve cell survival rates and inhibit lactate dehydrogenase release as well as apoptosis. CONCLUSIONS DNLP may protect the brain and neurons from hypoxic damage in mice with ischemic stroke by activating MCL-1 and downregulating miR-134, providing a new therapeutic target for ischemic stroke.
Collapse
|