1
|
Qi Y, Zhu H, Chen Y, Zhang Y, Jin S, Xu X, Ma X, Chen L, Zhao M, Zhu H, Yan P. 4-Hydroxydictyolactone alleviates cerebral ischemia injury by regulating neuroinflammation and autophagy via AMPK signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156157. [PMID: 39427520 DOI: 10.1016/j.phymed.2024.156157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/02/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Cerebral ischemia (CI), a cerebrovascular disorder, is a major contributor to disability and mortality. Marine-derived compounds are an important source of new neuroprotective drug candidates. Xenicane-type diterpenes from brown algae of the genus Dictyota have exhibited potential neuroprotective effects against CI injury, attributed to their antioxidant properties. However, whether there are other underlying neuroprotective mechanisms of xenicane diterpenes against CI is still ambiguous. PURPOSE This study aims to elucidate the neuroprotective efficacy and mechanism of 4-hydroxydictyolactone (HDTL) in the treatment of CI. METHODS The LPS-induced BV2 cell model was used for anti-neuroinflammatory activity assay. Tandem Mass Tag (TMT)-based quantitative proteomics was employed to identify underlying mechanisms. The OGD/R-induced SH-SY5Y cell model and a MCAO mice model were used to assess the neuroprotective effect of HDTL against CI in vitro and in vivo. RESULTS HDTL reduced inflammation in LPS-stimulated BV2 cells by inhibiting the IKK/IκB/NF-κB pathway and by enhancing AMPK phosphorylation. Additionally, in SH-SY5Y cells treated with OGD/R, HDTL facilitated autophagy and reduced apoptosis. The neuroprotective properties of HDTL were abrogated in AMPK- silenced SH-SY5Y cells. In MCAO mice, HDTL ameliorated CI injury as evidenced by decreases in neurological deficit scores and cerebral infarction. HDTL also promoted autophagy and reduced apoptosis in vivo through both the AMPK/mTOR and IKK/IκB/NF-κB pathways. CONCLUSION HDTL exhibits neuroprotective effects through regulating the AMPK/mTOR and IKK/IκB/NF-κB pathways. These findings suggest that HDTL is a promising therapeutic candidate for CI treatment.
Collapse
Affiliation(s)
- Yu Qi
- School of Traditional Chinese Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Haoyun Zhu
- School of Traditional Chinese Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Yinqi Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Yuanlong Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Shengjie Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Xiao Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Xiaohong Ma
- School of Traditional Chinese Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Leiqing Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Min Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Haoru Zhu
- School of Traditional Chinese Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China.
| | - Pengcheng Yan
- School of Traditional Chinese Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China.
| |
Collapse
|
2
|
Yan Y, Li Z, Zhang S, Bai F, Jing Y, Huang F, Yu Y. Remote limb ischemic preconditioning alleviated spinal cord injury through inhibiting proinflammatory immune response and promoting the survival of spinal neurons. Spinal Cord 2024; 62:562-573. [PMID: 39154149 DOI: 10.1038/s41393-024-01015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 08/19/2024]
Abstract
STUDY DESIGN Experimental animal study. OBJECTIVES To investigate the protective effect of remote limb ischemia preconditioning (RLPreC) on traumatic spinal cord injury (SCI) and explore the underlying biological mechanisms using RNA sequencing. SETTING China Rehabilitation Science Institute; Beijing; China. METHODS spinal cord injury was induced in mice using a force of 0.7 N. RLPreC treatment was administered. Motor function, pain behavior, and gene expression were assessed. RESULTS RLPreC treatment significantly improved motor function and reduced pain-like behavior in SCI mice. RNA-Seq analysis identified 5247 differentially expressed genes (DEGs). GO analysis revealed enrichment of immune response, inflammatory signaling, and synaptic transmission pathways among these DEGs. KEGG analysis indicated suppression of inflammation and promotion of synapse-related pathways. CONCLUSIONS RLPreC is a promising therapeutic strategy for improving motor function and alleviating pain after traumatic SCI. RNA-Seq analysis provides insights into potential therapeutic targets and warrants further investigation.
Collapse
Affiliation(s)
- Yitong Yan
- China Rehabilitation Science Institute, Beijing, People's Republic of China
- China Rehabilitation Research Center, Beijing, People's Republic of China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, People's Republic of China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, People's Republic of China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Zihan Li
- China Rehabilitation Science Institute, Beijing, People's Republic of China
- China Rehabilitation Research Center, Beijing, People's Republic of China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, People's Republic of China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, People's Republic of China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Shuangyue Zhang
- China Rehabilitation Science Institute, Beijing, People's Republic of China
- China Rehabilitation Research Center, Beijing, People's Republic of China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, People's Republic of China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, People's Republic of China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Fan Bai
- China Rehabilitation Science Institute, Beijing, People's Republic of China
- China Rehabilitation Research Center, Beijing, People's Republic of China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, People's Republic of China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, People's Republic of China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Yingli Jing
- China Rehabilitation Science Institute, Beijing, People's Republic of China
- China Rehabilitation Research Center, Beijing, People's Republic of China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, People's Republic of China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, People's Republic of China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Fubiao Huang
- China Rehabilitation Research Center, Beijing, People's Republic of China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Yan Yu
- China Rehabilitation Science Institute, Beijing, People's Republic of China.
- China Rehabilitation Research Center, Beijing, People's Republic of China.
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, People's Republic of China.
- School of Rehabilitation Medicine, Capital Medical University, Beijing, People's Republic of China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.
| |
Collapse
|
3
|
Zheng T, Jiang T, Ma H, Zhu Y, Wang M. Targeting PI3K/Akt in Cerebral Ischemia Reperfusion Injury Alleviation: From Signaling Networks to Targeted Therapy. Mol Neurobiol 2024; 61:7930-7949. [PMID: 38441860 DOI: 10.1007/s12035-024-04039-1] [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: 11/01/2023] [Accepted: 02/09/2024] [Indexed: 09/21/2024]
Abstract
Ischemia/reperfusion (I/R) injury is a pathological event that results in reperfusion due to low blood flow to an organ. Cerebral ischemia is a common cerebrovascular disease with high mortality, and reperfusion is the current standard intervention. However, reperfusion may further induce cellular damage and dysfunction known as cerebral ischemia/reperfusion injury (CIRI). Currently, strategies for the clinical management of CIRI are limited, necessitating the exploration of novel and efficacious treatment modalities for the benefit of patients. PI3K/Akt signaling pathway is an important cellular process associated with the disease. Stimulation of the PI3K/Akt pathway enhances I/R injury in multiple organs such as heart, brain, lung, and liver. It stands as a pivotal signaling pathway crucial for diminishing cerebral infarction size and safeguarding the functionality of brain tissue after CIRI. During CIRI, activation of the PI3K/Akt pathway exhibits a protective effect on CIRI. Furthermore, activation of the PI3K/Akt pathway has the potential to augment the activity of antioxidant enzymes, resulting in a decrease in reactive oxygen species (ROS) and the associated oxidative stress. Meanwhile, PI3K/Akt plays a neuroprotective role by inhibiting inflammatory responses and apoptosis. For example, PI3K/Akt interacts with NF-κB, Nrf2, and MAPK signaling pathways to mitigate CIRI. This article is aimed to explore the pivotal role and underlying mechanism of PI3K/Akt in ameliorating CIRI and investigate the influence of ischemic preconditioning and post-processing, as well as the impact of pertinent drugs or activators targeting the PI3K/Akt pathway on CIRI. The primary objective is to furnish compelling evidence supporting the activation of PI3K/Akt in the context of CIRI, elucidating its mechanistic intricacies. By doing so, the paper aims to underscore the critical contribution of PI3K/Akt in mitigating CIRI, providing a theoretical foundation for considering the PI3K/Akt pathway as a viable target for CIRI treatment.
Collapse
Affiliation(s)
- Ting Zheng
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Taotao Jiang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Hongxiang Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Yanping Zhu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Manxia Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
4
|
Li X, Xu B, Long L, Li Y, Xiao X, Qiu S, Xu J, Tian LW, Wang H. Phelligridimer A enhances the expression of mitofusin 2 and protects against cerebral ischemia/reperfusion injury. Chem Biol Interact 2024; 398:111090. [PMID: 38825057 DOI: 10.1016/j.cbi.2024.111090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/16/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Mitochondrial dysfunction and endoplasmic reticulum (ER) stress play pivotal roles in the pathology of cerebral ischemia. In this study, we investigated whether phelligridimer A (PA), an active compound isolated from the medicinal and edible fungus Phellinus igniarius, ameliorates ischemic cerebral injury by restoring mitochondrial function and restricting ER stress. An in vitro cellular model of ischemic stroke-induced neuronal damage was established by exposing HT-22 neuronal cells to oxygen-glucose deprivation/reoxygenation (OGD/R). An in vivo animal model was established in rats subjected to middle cerebral artery occlusion/reperfusion (MCAO/R). The results showed that PA (1-10 μM) dose-dependently increased HT-22 cell viability, reduced OGD/R-induced lactate dehydrogenase release, and reversed OGD/R-induced apoptosis. PA reduced OGD/R-induced accumulation of reactive oxygen species, restored mitochondrial membrane potential, and increased ATP levels. Additionally, PA reduced the expression of the 78-kDa glucose-regulated protein (GRP78) and the phosphorylation of inositol-requiring enzyme-1α (p-IRE1α) and eukaryotic translation-initiation factor 2α (p-eIF2α). PA also inhibited the activation of the mitogen-activated protein kinase (MAPK) pathway in the OGD/R model. Moreover, treatment with PA restored the expression of mitofusin 2 (Mfn-2), a protein linking mitochondria and ER. The silencing of Mfn-2 abolished the protective effects of PA. The results from the animal study showed that PA (3-10 mg/kg) significantly reduced the volume of cerebral infarction and neurological deficits, which were accompanied by an increased level of Mfn-2, and decreased activation of the ER stress in the penumbra of the ipsilateral side after MCAO/R in rats. Taken together, these results indicate that PA counteracts cerebral ischemia-induced injury by restoring mitochondrial function and reducing ER stress. Therefore, PA might be a novel protective agent to prevent ischemia stroke-induced neuronal injury.
Collapse
Affiliation(s)
- Xing Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bingtian Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lu Long
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yuting Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xuan Xiao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuqin Qiu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiangping Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, 510515, China
| | - Li-Wen Tian
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Haitao Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Li X, Qiao M, Zhou Y, Peng Y, Wen G, Xie C, Zhang Y. Modulating the RPS27A/PSMD12/NF-κB pathway to control immune response in mouse brain ischemia-reperfusion injury. Mol Med 2024; 30:106. [PMID: 39039432 PMCID: PMC11265174 DOI: 10.1186/s10020-024-00870-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/01/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Investigating immune cell infiltration in the brain post-ischemia-reperfusion (I/R) injury is crucial for understanding and managing the resultant inflammatory responses. This study aims to unravel the role of the RPS27A-mediated PSMD12/NF-κB axis in controlling immune cell infiltration in the context of cerebral I/R injury. METHODS To identify genes associated with cerebral I/R injury, high-throughput sequencing was employed. The potential downstream genes were further analyzed using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein-Protein Interaction (PPI) analyses. For experimental models, primary microglia and neurons were extracted from the cortical tissues of mouse brains. An in vitro cerebral I/R injury model was established in microglia using the oxygen-glucose deprivation/reoxygenation (OGD/R) technique. In vivo models involved inducing cerebral I/R injury in mice through the middle cerebral artery occlusion (MCAO) method. These models were used to assess neurological function, immune cell infiltration, and inflammatory factor release. RESULTS The study identified RPS27A as a key player in cerebral I/R injury, with PSMD12 likely acting as its downstream regulator. Silencing RPS27A in OGD/R-induced microglia decreased the release of inflammatory factors and reduced neuron apoptosis. Additionally, RPS27A silencing in cerebral cortex tissues mediated the PSMD12/NF-κB axis, resulting in decreased inflammatory factor release, reduced neutrophil infiltration, and improved cerebral injury outcomes in I/R-injured mice. CONCLUSION RPS27A regulates the expression of the PSMD12/NF-κB signaling axis, leading to the induction of inflammatory factors in microglial cells, promoting immune cell infiltration in brain tissue, and exacerbating brain damage in I/R mice. This study introduces novel insights and theoretical foundations for the treatment of nerve damage caused by I/R, suggesting that targeting the RPS27A and downstream PSMD12/NF-κB signaling axis for drug development could represent a new direction in I/R therapy.
Collapse
Affiliation(s)
- Xiaocheng Li
- Key Laboratory of Clinical Genetics, Affiliated Hospital of Chengdu University & College of Food and Biological Engineering, Chengdu, 610081, P. R. China
| | - Ming Qiao
- Department of Critical Medicine, The People's Hospital of Renshou County, Meishan, 620500, P. R. China
| | - Yan Zhou
- Department of Radiation Protection Medicine, Faculty of Preventive Medicine, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Yan Peng
- Department of Critical Medicine, The People's Hospital of Renshou County, Meishan, 620500, P. R. China
| | - Gang Wen
- Department of Critical Medicine, The People's Hospital of Renshou County, Meishan, 620500, P. R. China
| | - Chenchen Xie
- Department of Neurology, Affiliated Hospital of Chengdu University, Chengdu, 610082, P. R. China
| | - Yamei Zhang
- Key Laboratory of Clinical Genetics, Affiliated Hospital of Chengdu University, No. 82, North Section 2, 2nd Ring Road, Chengdu, Sichuan, 610081, P. R. China.
| |
Collapse
|
6
|
Zhou X, Luo X. Shionone relieves oxygen-glucose deprivation/reoxygenation induced SH-SY5Y cells injury by inhibiting the p38 MAPK/NF-κB pathway. J Cardiothorac Surg 2024; 19:435. [PMID: 38997740 PMCID: PMC11241947 DOI: 10.1186/s13019-024-02938-x] [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: 11/14/2023] [Accepted: 06/16/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (I/R) can affect patient outcomes and can even be life-threatening. This study aimed to explore the role of Shionone in cerebral I/R and reveal its mechanism of action through the cerebral I/R in vitro model. METHODS SH-SY5Y cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to induce cerebral I/R in vitro model. SH-SY5Y cells were treated with different concentrations of Shionone. Cell counting kit-8 and flow cytometry assays were used to detect cell viability and apoptosis levels. The levels of superoxide dismutase, catalase, and malondialdehyde were determined using their corresponding kits to examine the level of oxidative stress. The inflammation response was detected by IL-6, IL-1β, and TNF-α levels, using enzyme-linked-immunosorbent-assay. RT-qPCR was performed to measure the mRNA levels of p38 and NF-κB. Western blotting was used to quantify the apoptosis-related proteins and p38MAPK/NF-κB signaling pathway proteins. RESULTS Shionone exhibited no toxic effects on SH-SY5Y cells. Shionone inhibited OGD/R-induced cell apoptosis, improved the inflammatory response caused by OGD/R, and reduced the level of oxidative stress in cells. Western blot assay results showed that Shionone alleviated OGD/R-induced injury by inhibiting the activity of the p38 MAPK/NF-κB signaling pathway. The p38/MAPK agonist P79350 reversed the beneficial effects of Shionone. CONCLUSION Shionone alleviates cerebral I/R and may thus be a novel therapeutic strategy for treating cerebral I/R.
Collapse
Affiliation(s)
- Xiaoli Zhou
- Department of Neurology, The Affiliated Hospital of Hubei University of Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Xueting Luo
- Department of Cardiovascular Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, No. 856 Luoyu Road, Hongshan District, Wuhan, 430061, China.
| |
Collapse
|
7
|
Xu B, Wu H, Guo W, Hussain SA, Wang T. Voacangine mitigates oxidative stress and neuroinflammation in middle cerebral artery occlusion-induced cerebral ischemia/reperfusion injury by averting the NF-κBp65/MAPK signaling pathways in rats. ENVIRONMENTAL TOXICOLOGY 2024; 39:4004-4013. [PMID: 38606816 DOI: 10.1002/tox.24274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/18/2024] [Accepted: 03/24/2024] [Indexed: 04/13/2024]
Abstract
Ischemic stroke is a leading cause of human mortality. Cerebral ischemia-reperfusion injury (CI/RI) is a primary cause of stroke. Ischemia-reperfusion (I/R) resulting in oxidative stress and inflammatory events may lead to severe neuronal impairments. Thus, anti-oxidative and anti-inflammatory mediators that can alleviate post-I/R neuronal injuries are required for the treatment of CI/RI. An alkaloid, voacangine (VCG) is a recognized antioxidant, anti-inflammatory, and anticancer agent. Hence, the current study intended to explore the neuroprotective potential and the principal mechanisms of VCG in CI/RI. The experimental rats were divided into four sets: control, I/R-induced, I/R + VCG (2.5 mg/kg), I/R + VCG (5 mg/kg). CI/RI was induced by implanting a thread into the middle cerebral artery occlusion (MCAO) model. Brain damages were assessed on the basis of brain edema, brain infarct volume, neurological deficit score, histopathology, oxidative stress, and neuroinflammation. Results revealed that VCG inhibited the triggering of NLRP3 inflammasome, pro-inflammatory cytokines, lipid peroxidation, but enhanced the antioxidant status in MCAO rats. Furthermore, VCG treatment averted brain damage by I/R, neuroinflammation, and oxidative stress by suppressing NF-κBp65/MAPK pathways. The results of the study provide pertinent insights pertaining to the role of VCG as a potential neuroprotective agent against ischemic stroke.
Collapse
Affiliation(s)
- Bo Xu
- Department of General Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Hua Wu
- Shaanxi Provincial Center for Diseases Control and Prevention, Xi'an, China
| | - Wei Guo
- Department of General Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Tian Wang
- Department of Geratology, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
8
|
Xu L, Mi Y, Meng Q, Liu Y, Wang F, Zhang G, Liu Y, Chen G, Hou Y. Anti-inflammatory effects of quinolinyl analog of resveratrol targeting TLR4 in MCAO/R ischemic stroke rat model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155344. [PMID: 38493721 DOI: 10.1016/j.phymed.2024.155344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/25/2023] [Accepted: 01/07/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Among adults, stroke is the main causes of mortality and permanent disability. Neuroinflammation is one of the main causes of stoke-mediated neuronal death. Our previous study revealed that (E)-5-(2-(Quinolin-4-yl) vinyl) benzene-1, 3-diol (RV01), a quinolinyl analog of resveratrol, inhibits microglia-induced neuroinflammation and safeguards neurons from inflammatory harm. The preventive role of RV01 in ischemic stroke and its underlying cellular mechanisms and molecular targets remain poorly understood. PURPOSE To investigate whether RV01 alleviates ischemia-reperfusion (I/R) injury by inhibiting microglia-mediated neuroinflammation and determine the potential molecular mechanisms and targets by which RV01 inhibits the I/R-mediated microglia activation. METHODS Rat middle cerebral artery occlusion and reperfusion (MCAO/R) and BV-2 or primary microglial cells oxygen-glucose deprivation and reperfusion (OGD/R) models were established. The neurological behavior scores, 2, 3, 5-triphenyl tetrazolium chloride staining and immunofluorescence were used to detect the neuroprotective effect of RV01 in the MCAO/R rats. In addition, the mRNA expression levels of IL-6, TNF-α, and IL-1β were detected to reveal the antineuroinflammatory effect of RV01. Moreover, a western blot assay was performed to explore the protein expression changes in NF-κB-mediated neuroinflammation. Finally, we identified TLR4 as an RV01 target through molecular docking, drug sensitivity target stability analysis, cellular thermal shift analysis, and surface plasmon resonance techniques. RESULTS RV01 reduced the infarct volume and neurological deficits, increased the rotarod duration, and decreased the number of rightward deflections in the MCAO/R rats. RV01 inhibited the NF-κB signaling pathway in vitro and in vivo, as demonstrated by the reduction in the transcription factor p65-mediated expression of several inflammatory factors including IL-6, TNF-α, and IL-1β. Further studies showed that its protective effect was associated with targeting the TLR4 protein. Notably, the anti-inflammatory effect of RV01 was markedly reinforced by the TLR4 knockdown, but inhibited by the overexpression of TLR4. Results revealed that the conditioned medium derived from the RV01-treated BV-2 cells significantly decreased the OGD/R-mediated neuronal damage. CONCLUSION Our results are the first to reveal the protective effects of RV01 on cerebral ischemia, depending on its inhibitory effect on the NF-κB pathway by targeting TLR4. RV01 could be a potential protective agent in ischemic stroke treatment.
Collapse
Affiliation(s)
- Libin Xu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yan Mi
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Qingqi Meng
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yeshu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Feng Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Guijie Zhang
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Key Laboratory of Vascular Biology, Science and Research Center, Shenyang Medical College, Shenyang, China.
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China.
| |
Collapse
|
9
|
Zhu J, Park S, Kim SH, Kim CH, Jeong KH, Kim WJ. Sirtuin 3 regulates astrocyte activation by reducing Notch1 signaling after status epilepticus. Glia 2024; 72:1136-1149. [PMID: 38406970 DOI: 10.1002/glia.24520] [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: 11/14/2023] [Revised: 01/31/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024]
Abstract
Sirtuin3 (Sirt3) is a nicotinamide adenine dinucleotide enzyme that contributes to aging, cancer, and neurodegenerative diseases. Recent studies have reported that Sirt3 exerts anti-inflammatory effects in several neuropathophysiological disorders. As epilepsy is a common neurological disease, in the present study, we investigated the role of Sirt3 in astrocyte activation and inflammatory processes after epileptic seizures. We found the elevated expression of Sirt3 within reactive astrocytes as well as in the surrounding cells in the hippocampus of patients with temporal lobe epilepsy and a mouse model of pilocarpine-induced status epilepticus (SE). The upregulation of Sirt3 by treatment with adjudin, a potential Sirt3 activator, alleviated SE-induced astrocyte activation; whereas, Sirt3 deficiency exacerbated astrocyte activation in the hippocampus after SE. In addition, our results showed that Sirt3 upregulation attenuated the activation of Notch1 signaling, nuclear factor kappa B (NF-κB) activity, and the production of interleukin-1β (IL1β) in the hippocampus after SE. By contrast, Sirt3 deficiency enhanced the activity of Notch1/NF-κB signaling and the production of IL1β. These findings suggest that Sirt3 regulates astrocyte activation by affecting the Notch1/NF-κB signaling pathway, which contributes to the inflammatory response after SE. Therefore, therapies targeting Sirt3 may be a worthy direction for limiting inflammatory responses following epileptic brain injury.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soojin Park
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| | - Chul Hoon Kim
- Department of Pharmacology, Brain Korea 21 Project, Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Hoon Jeong
- Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won-Joo Kim
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Chen H, Zhang T, Yan S, Zhang S, Fu Q, Xiong C, Zhou L, Ma X, Wang R, Chen G. Protective effects of the bioactive peptide from maggots against skin flap ischemia‒reperfusion injury in rats. Heliyon 2024; 10:e29874. [PMID: 38694094 PMCID: PMC11058300 DOI: 10.1016/j.heliyon.2024.e29874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024] Open
Abstract
Ischemia‒reperfusion (I/R) injury is a frequently observed complication after flap surgery, and it affects skin flap survival and patient prognosis. Currently, there are no proven safe and effective treatment options to treat skin flap I/R injury. Herein, the potential efficacies of the bioactive peptide from maggots (BPM), as well as its underlying mechanisms, were explored in a rat model of skin flap I/R injury and LPS- or H2O2-elicited RAW 264.7 cells. We demonstrated that BPM significantly ameliorated the area of flap survival, and histological changes in skin tissue in vivo. Furthermore, BPM could markedly restore or enhance Nrf2 and HO-1 levels, and suppress the expression of pro-inflammatory cytokines, including TLR4, p-IκB, NFκB p65, p-p65, IL-6, and TNF-α in I/R-injured skin flaps. In addition, BPM treatment exhibited excellent biocompatibility with an adequate safety profile, while it exhibited superior ROS-scavenging ability and the upregulation of antioxidant enzymes in vitro. Mechanistically, the above benefits related to BPM involved the activation of Nrf2/HO-1 and suppression of TLR4/NF-κB pathway. Taken together, this study may provide a scientific basis for the potential therapeutic effect of BPM in the prevention of skin flap I/R injury and other related diseases.
Collapse
Affiliation(s)
- Hao Chen
- Department of Plastic Surgery, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Tianqi Zhang
- Department of Plastic Surgery, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Su Yan
- Department of Plastic Surgery, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Shan Zhang
- Department of Plastic Surgery, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Qiuyue Fu
- Department of Plastic Surgery, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Chuchu Xiong
- Department of Plastic Surgery, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Lina Zhou
- Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, PR China
| | - Xiao Ma
- Yixing Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Yixing, Jiangsu, PR China
| | - Rong Wang
- College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, PR China
| | - Gang Chen
- Department of Plastic Surgery, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, PR China
| |
Collapse
|
11
|
Wu Q, Li Y, Ye R, Wang H, Ge Y. Velvet antler polypeptide (VAP) protects against cerebral ischemic injury through NF-κB signaling pathway in vitro. J Stroke Cerebrovasc Dis 2024; 33:107666. [PMID: 38423152 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107666] [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: 11/16/2023] [Revised: 02/06/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE Velvet antler polypeptide (VAP) has been shown to play important roles in the immune and nervous systems. The purpose of this study was to investigate the protective effects of VAP on cerebral ischemic injury with the involvement of NF-κB signaling pathway in vitro. MATERIALS AND METHODS PC-12 cells stimulated by oxygen-glucose deprivation/reperfusion (OGD/R) was used to mimic cerebral ischemic injury in vitro. The levels of ROS, SOD, and intracellular concentrations of Ca2+ were measured by the relevant kits. Meanwhile, the expressions of inflammatory cytokines (IL-6, IL-1β, and TNF-α) were determined by ELISA kit assay. In addition, MTT, EdU, and flow cytometry assays were used to measure the cell proliferation and apoptosis. Besides which, the related proteins of NF-κB signaling pathway were measured by western blotting assay. RESULTS VAP alleviated cerebral ischemic injury by reducing OGD/R-induced oxidative stress, inflammation, and apoptosis in PC-12 cells in a time dependent manner. Mechanistically, VAP inhibited the levels of p-p65 and p-IkB-α in a time dependent manner, which was induced by OGD/R operation. Moreover, NF-κB agonist diprovocim overturned the suppression effects of VAP on OGD/R-induced oxidative stress, inflammation, and apoptosis in PC-12 cells. CONCLUSIONS The results demonstrate that VAP may alleviate cerebral ischemic injury by suppressing the activation of NF-κB signaling pathway.
Collapse
Affiliation(s)
- Qian Wu
- Physical Examination Center, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430074, Hubei Province, China
| | - Yutao Li
- Physical Examination Center, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430074, Hubei Province, China.
| | - Ru Ye
- Physical Examination Center, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430074, Hubei Province, China
| | - Hui Wang
- Physical Examination Center, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430074, Hubei Province, China
| | - Ying Ge
- Physical Examination Center, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430074, Hubei Province, China
| |
Collapse
|
12
|
Li Z, Su H, Lin G, Wang K, Huang Y, Wen Y, Luo D, Hou Y, Cao X, Weng J, Lin D, Wang L, Li X. Transplantation of MiR-28-5p-Modified BMSCs Promotes Functional Recovery After Spinal Cord Injury. Mol Neurobiol 2024; 61:2197-2214. [PMID: 37864767 DOI: 10.1007/s12035-023-03702-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/06/2023] [Indexed: 10/23/2023]
Abstract
Traumatic spinal cord injury (TSCI) is a prevalent central nervous system condition that imposes a significant burden on both families and society, affecting more than 2 million people worldwide. Recently, there has been increasing interest in bone marrow mesenchymal stem cell (BMSC) transplantation as a promising treatment for spinal cord injury (SCI) due to their accessibility and low immunogenicity. However, the mere transplantation of BMSCs has limited capacity to directly participate in the repair of host spinal cord nerve function. MiR-28-5p, identified as a key differentially expressed miRNA in spinal cord ischemia-reperfusion injury, exhibits differential expression and regulation in various neurological diseases. Nevertheless, its involvement in this process and its specific regulatory mechanisms in SCI remain unclear. Therefore, this study aimed to investigate the potential mechanisms through which miR-28-5p promotes the neuronal differentiation of BMSCs both in vivo and in vitro. Our results indicate that miR-28-5p may directly target Notch1, thereby facilitating the neuronal differentiation of BMSCs in vitro. Furthermore, the transplantation of lentivirus-mediated miR-28-5p-overexpressed BMSCs into SCI rats effectively improved footprint tests and Basso, Beattie, and Bresnahan (BBB) scores, ameliorated histological morphology (hematoxylin-eosin [HE] and Nissl staining), promoted axonal regeneration (MAP2 and growth-associated protein 43 [GAP43]), and facilitated axonal remyelination (myelin basic protein [MBP]). These findings may suggest that miR-28-5p-modified BMSCs could serve as a therapeutic target to enhance the behavioral and neurological recovery of SCI rats.
Collapse
Affiliation(s)
- Zhen Li
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Haitao Su
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Guandai Lin
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Kai Wang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Yongming Huang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Yaqian Wen
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Dan Luo
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Yu Hou
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Xuewei Cao
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Jiaxian Weng
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Dingkun Lin
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Le Wang
- Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-Sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, 510080, Guangdong, China.
| | - Xing Li
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China.
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, 510120, Guangdong, China.
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| |
Collapse
|
13
|
Liu Y, Yu R, Wang X, Chen Y, Yin T, Gao Q, Sun L, Zheng Z. Research progress of the effective active ingredients of Astragalus mongholicus in the treatment of diabetic peripheral neuropathy. Biomed Pharmacother 2024; 173:116350. [PMID: 38430632 DOI: 10.1016/j.biopha.2024.116350] [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: 12/06/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
Diabetic peripheral neuropathy (DPN) is one of the most prevalent consequences of diabetes, with a high incidence and disability rate. The DPN's pathogenesis is extremely complex and yet to be fully understood. Persistent high glucose metabolism, nerve growth factor deficiency, microvascular disease, oxidative stress, peripheral nerve cell apoptosis, immune factors, and other factors have been implicated in the pathogenesis of DPN. Astragalus mongholicus is a commonly used plant used to treat DPN in clinical settings. Its rich chemical components mainly include Astragalus polysaccharide, Astragalus saponins, Astragalus flavones, etc., which play a vital role in the treatment of DPN. This review aimed to summarize the pathogenesis of DPN and the studies on the mechanism of the effective components of Astragalus mongholicus in treating DPN. This is of great significance for the effective use of Chinese herbal medicine and the promotion of its status and influence on the world.
Collapse
Affiliation(s)
- Yulian Liu
- Department of Rehabilitation Medicine,The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Runyuan Yu
- Department of Rehabilitation Medicine,The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Xiaoyu Wang
- Department of Rehabilitation Medicine,The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Yuexia Chen
- Department of Skills Training Center,The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Tao Yin
- Department of Rehabilitation Medicine,The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Qiang Gao
- Department of Rehabilitation Medicine,The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Limin Sun
- Department of Rehabilitation Medicine,The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Zuncheng Zheng
- Department of Rehabilitation Medicine,The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China.
| |
Collapse
|
14
|
Zhang L, Guo Q, An R, Shen S, Yin L. In vitro ischemic preconditioning mediates the Ca 2+/CaN/NFAT pathway to protect against oxygen-glucose deprivation-induced cellular damage and inflammatory responses. Brain Res 2024; 1826:148736. [PMID: 38141801 DOI: 10.1016/j.brainres.2023.148736] [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: 11/06/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Oxygen-glucose deprivation (OGD) is a critical model for studying hypoxic-ischemic cerebrovascular disease in vitro. This paper is to investigate the protection of OGD-induced cellular damage and inflammatory responses by OGD preconditioning in vitro, to provide a theoretical basis for OGD preconditioning to improve the prevention and prognosis of ischemic stroke. OGD or OGD preconditioning model was established by culturing the PC12 cell line in vitro, followed by further adding A23187 (calcium ion carrier) or CsA (calcium ion antagonist). Cell viability was detected by MTT, apoptosis by Hoechst 33,258 staining, the levels of TNF-α and IL-1β mRNA by RT-qPCR and ELISA, and the levels of CaN, NFAT, COX-2 by RT-qPCR and Western blot. Cell viability was decreased, and apoptosis, inflammatory cytokines, and CaN, NFAT, and COX-2 levels were notably increased upon OGD, while OGD pretreatment significantly increased cell viability and decreased apoptosis, inflammation, and the Ca2+/CaN/NFAT pathway. Treatment with A23187 decreased cell viability, promoted apoptosis, and significantly increased TNF-α, IL-1β, CaN, NFAT, and COX-2 levels, while CsA treatment reduced the opposite results. In vitro OGD preconditioning mediates the Ca2+/CaN/NFAT pathway to protect against OGD-induced cellular damage and inflammatory responses.
Collapse
Affiliation(s)
- Lihong Zhang
- Department of Neurointervention and Neurocritical Care, Dalian Central Hospital, Affiliated to Dalian University of Technology, Dalian, China; Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian 116023, Liaoning, China
| | - Qingzi Guo
- Deprtment of Cardiothoracic Surgery, Royal Stoke University Hospital, Stoke-on-Trent ST4 6QG, UK
| | - Ran An
- Department of Neurology, Taihe Hospital, Shiyan 442099, Hubei, China
| | - Shuhan Shen
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121012, Liaoning, China
| | - Lin Yin
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian 116023, Liaoning, China.
| |
Collapse
|
15
|
Han Q, Wang F. Electroacupuncture at GB20 improves cognitive ability and synaptic plasticity via the CaM-CaMKII-CREB signaling pathway following cerebral ischemia-reperfusion injury in rats. Acupunct Med 2024; 42:23-31. [PMID: 38126262 DOI: 10.1177/09645284231202805] [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] [Indexed: 12/23/2023]
Abstract
BACKGROUND This study aimed to investigate the effects of electroacupuncture (EA) on cognitive recovery and synaptic remodeling in a rat model of middle cerebral artery occlusion (MCAO) followed by reperfusion and explore the possible mechanism. METHOD Focal cerebral ischemia was modeled in healthy adult Sprague-Dawley rats by MCAO. The MCAO rats were classified into four groups: sham, MCAO, MCAO + GB20 (receiving EA at GB20) and MCAO + NA (receiving EA at a "non-acupoint" location not corresponding to any traditional acupuncture point location about 10 mm above the iliac crest). Neurological deficit scores and behavior were assessed before and during the treatment. After intervention for 7 days, the hippocampus was dissected to analyze growth-associated protein (GAP)-43, synaptophysin (SYN) and postsynaptic density protein (PSD)-95 expression levels by Western blotting. Bioinformatic analysis and primary hippocampal neurons with calcium-voltage gated channel subunit alpha 1B (CACNA1B) gene overexpression were used to screen the target genes for EA against MCAO. RESULTS Significant amelioration of neurological deficits and learning/memory were found in MCAO + GB20 rats compared with MCAO or MCAO + NA rats. Protein levels of GAP-43, SYN and PSD-95 were significantly improved in MCAO + GB20-treated rats together with an increase in the number of synapses in the hippocampal CA1 region. CACNA1B appeared to be a target gene of EA in MCAO. There were increased mRNA levels of CACNA1B, calmodulin (CaM), Ca2+/calmodulin-dependent protein kinase type II (CaMKII) and cyclic adenosine monophosphate response element binding (CREB) and increased phosphorylation of CaM, CaMKII and CREB in the hippocampal region in MCAO + GB20 versus MCAO and MCAO + NA groups. CACNA1B overexpression modulated expression of the CaM-CaMKII-CREB axis. CONCLUSION EA treatment at GB20 may ameliorate the negative effects of MCAO on cognitive function in rats by enhancing synaptic plasticity. EA treatment at GB20 may exert this neuroprotective effect by regulating the CACNA1B-CaM-CaMKII-CREB axis.
Collapse
Affiliation(s)
- Qing Han
- Department of Traditional Chinese Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| | - Feng Wang
- Department of Traditional Chinese Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
16
|
Lin G, Jiang H, Zhang Z, Ning L, Zhang W, Peng L, Xu S, Sun W, Tao S, Zhang T, Tang L. Molecular mechanism of NR4A1/MDM2/P53 signaling pathway regulation inducing ferroptosis in renal tubular epithelial cells involved in the progression of renal ischemia-reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166968. [PMID: 38008232 DOI: 10.1016/j.bbadis.2023.166968] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/26/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Revealing the possible molecular mechanism of the NR4A1 (nuclear receptor subfamily 4 group A member 1)-MDM2 (MDM2 proto-oncogene)-P53 (tumor protein p53) signaling pathway that induces ferroptosis in renal tubular epithelial cells. Renal ischemia-reperfusion injury (RIRI) -related datasets were obtained from the GEO database. Differentially expressed genes in RIRI were analyzed using R language, intersected with RIRI-related genes in the GeneCard database, and retrieved from the literature to finally obtain differential ferroptosis-related genes. An in vitro cell model of RIRI was constructed using mouse renal cortical proximal tubule epithelial cells (mRTEC cells) treated with hypoxia-reoxygenation (H/R). Bioinformatic analysis showed that NR4A1 may be involved in RIRI through the induction of ferroptosis; in addition, we predicted through online databases that the downstream target gene of NR4A1, MDM2, could be targeted and regulated by ChIP and dual luciferase assays, and that NR4A1 could prevent MDM2 by inhibiting it, and NR4A1 was able to promote ferroptosis by inhibiting the ubiquitinated degradation of P53. NR4A1 expression was significantly increased in mRTEC cells in the hypoxia/reoxygenation model, and the expression of ferroptosis-related genes was increased in vitro experiments. NR4A1 reduces the ubiquitinated degradation of P53 by targeting the inhibition of MDM2 expression, thereby inducing ferroptosis and ultimately exacerbating RIRI by affecting the oxidative respiration process in mitochondria and producing oxidized lipids. This study presents a novel therapeutic approach for the clinical treatment of renal ischemia-reperfusion injury by developing drugs that inhibit NR4A1 to alleviate kidney damage caused by renal ischemia-reperfusion.
Collapse
Affiliation(s)
- Guangzheng Lin
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Heng Jiang
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhihui Zhang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Ling Ning
- Department of Infectious Diseases, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei 230000, PR China
| | - Wenbo Zhang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Longfei Peng
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Shen Xu
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Wei Sun
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Sha Tao
- Second School of Clinical Medicine, Anhui Medical University, Hefei 230601, PR China
| | - Tao Zhang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Liang Tang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| |
Collapse
|
17
|
Zhang M, Liu Q, Meng H, Duan H, Liu X, Wu J, Gao F, Wang S, Tan R, Yuan J. Ischemia-reperfusion injury: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:12. [PMID: 38185705 PMCID: PMC10772178 DOI: 10.1038/s41392-023-01688-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemia-reperfusion (I/R) injury paradoxically occurs during reperfusion following ischemia, exacerbating the initial tissue damage. The limited understanding of the intricate mechanisms underlying I/R injury hinders the development of effective therapeutic interventions. The Wnt signaling pathway exhibits extensive crosstalk with various other pathways, forming a network system of signaling pathways involved in I/R injury. This review article elucidates the underlying mechanisms involved in Wnt signaling, as well as the complex interplay between Wnt and other pathways, including Notch, phosphatidylinositol 3-kinase/protein kinase B, transforming growth factor-β, nuclear factor kappa, bone morphogenetic protein, N-methyl-D-aspartic acid receptor-Ca2+-Activin A, Hippo-Yes-associated protein, toll-like receptor 4/toll-interleukine-1 receptor domain-containing adapter-inducing interferon-β, and hepatocyte growth factor/mesenchymal-epithelial transition factor. In particular, we delve into their respective contributions to key pathological processes, including apoptosis, the inflammatory response, oxidative stress, extracellular matrix remodeling, angiogenesis, cell hypertrophy, fibrosis, ferroptosis, neurogenesis, and blood-brain barrier damage during I/R injury. Our comprehensive analysis of the mechanisms involved in Wnt signaling during I/R reveals that activation of the canonical Wnt pathway promotes organ recovery, while activation of the non-canonical Wnt pathways exacerbates injury. Moreover, we explore novel therapeutic approaches based on these mechanistic findings, incorporating evidence from animal experiments, current standards, and clinical trials. The objective of this review is to provide deeper insights into the roles of Wnt and its crosstalk signaling pathways in I/R-mediated processes and organ dysfunction, to facilitate the development of innovative therapeutic agents for I/R injury.
Collapse
Affiliation(s)
- Meng Zhang
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
| | - Qian Liu
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hui Meng
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hongxia Duan
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Xin Liu
- Second Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fei Gao
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Rubin Tan
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China.
| |
Collapse
|
18
|
Baranova K, Nalivaeva N, Rybnikova E. Neuroadaptive Biochemical Mechanisms of Remote Ischemic Conditioning. Int J Mol Sci 2023; 24:17032. [PMID: 38069355 PMCID: PMC10707673 DOI: 10.3390/ijms242317032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
This review summarizes the currently known biochemical neuroadaptive mechanisms of remote ischemic conditioning. In particular, it focuses on the significance of the pro-adaptive effects of remote ischemic conditioning which allow for the prevention of the neurological and cognitive impairments associated with hippocampal dysregulation after brain damage. The neuroimmunohumoral pathway transmitting a conditioning stimulus, as well as the molecular basis of the early and delayed phases of neuroprotection, including anti-apoptotic, anti-oxidant, and anti-inflammatory components, are also outlined. Based on the close interplay between the effects of ischemia, especially those mediated by interaction of hypoxia-inducible factors (HIFs) and steroid hormones, the involvement of the hypothalamic-pituitary-adrenocortical system in remote ischemic conditioning is also discussed.
Collapse
Affiliation(s)
| | | | - Elena Rybnikova
- I. P. Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia; (K.B.); (N.N.)
| |
Collapse
|
19
|
Wang M, Wei G, Gu S, Huo Z, Han X. A Machine Learning-Based Classification of Immunogenic Cell Death Regulators and Characterisation of Immune Microenvironment in Acute Ischemic Stroke. Int J Clin Pract 2023; 2023:9930172. [PMID: 38020537 PMCID: PMC10663090 DOI: 10.1155/2023/9930172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Immunogenic cell death (ICD) regulators exert a crucial part in quite a few in numerous biological processes. This study aimed to determine the function and diagnostic value of ICD regulators in acute ischemic stroke (AIS). 31 significant ICD regulators were identified from the gene expression omnibus (GEO) database in this work (the combination of the GSE16561 dataset and the GSE37587 dataset in the comparison of non-AIS and AIS patients). The random forest model was applied and 15 potential ICD regulators were screened to forecast the probability of AIS. A nomogram, on the basis of 11 latent ICD regulators, was performed. The resolution curve analysis indicated that patients can gain benefits from the nomogram. The consensus clustering approach was applied, and AIS patients were divided into 2 ICD clusters (cluster A and cluster B) based on the identified key ICD regulatory factors. To quantify the ICD pattern, 181 ICD-related dissimilarly expressed genes (DEGs) were selected for further investigation. The expression levels of NFKB1, NFKB2, and PARP1 were greater in gene cluster A than in gene cluster B. In conclusion, ICD regulators exerted a crucial part in the progress of AIS. The investigation made by us on ICD patterns perhaps informs prospective immunotherapeutic methods for AIS.
Collapse
Affiliation(s)
- Mengying Wang
- Department of Anesthesiology, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guolian Wei
- Department of Neurosurgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Shaorui Gu
- Department of Thoracic Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, Shanghai 200065, China
| | - Zhengyuan Huo
- Department of Neurosurgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Xue Han
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200065, China
| |
Collapse
|
20
|
Chen Y, Pang J, Ye L, Zhang Z, Lin S, Lin N, Lee TH, Liu H. Disorders of the central nervous system: Insights from Notch and Nrf2 signaling. Biomed Pharmacother 2023; 166:115383. [PMID: 37643483 DOI: 10.1016/j.biopha.2023.115383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
The functional complexity of the central nervous system (CNS) is unparalleled in living organisms. It arises from neural crest-derived cells that migrate by the exact route, leading to the formation of a complex network of neurons and glial cells. Recent studies have shown that novel crosstalk exists between the Notch1 and Nrf2 pathways and is associated with many neurological diseases. The Notch1-Nrf2 axis may act on nervous system development, and the molecular mechanism has recently been reported. In this review, we summarize the essential structure and function of the CNS. The significance of interactions between signaling pathways and between developmental processes like proliferation, apoptosis and migration in ensuring the correct development of the CNS is also presented. We primarily focus on research concerning possible mechanism of interaction between Notch1 and Nrf2 and the functions of Notch1-Nrf2 in neurons. There may be a direct interaction between Notch1 and NRF2, which is closely related to the crosstalk that occurs between them. The significance and potential applications of the Notch1-Nrf2 axis in abnormal development of the nervous system are been highlighten. We also discuss the molecular mechanisms by which the Notch1-Nrf2 axis controls the apoptosis, antioxidant pathway and differentiation of neurons to modulate the development of the nervous system. This information will lead to a better understanding of Notch1-Nrf2 axis signaling pathways in the nervous system and may facilitate the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Yuwen Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jiao Pang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Lu Ye
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Zhentao Zhang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Suijin Lin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Na Lin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Tae Ho Lee
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Hekun Liu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China.
| |
Collapse
|
21
|
Wei L, Li X, Wei Q, Chen L, Xu L, Zhou P. Oxidative Stress-mediated Sprouty-related Protein with an EVH1 Domain 1 Down-regulation Contributes to Resisting Oxidative Injury in Microglia. Neuroscience 2023; 526:13-20. [PMID: 37343716 DOI: 10.1016/j.neuroscience.2023.06.012] [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: 02/22/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023]
Abstract
Microglia play an ambiguous role in injury or repair after ischemia-reperfusion, and the induced oxidative stress serves as an important signal, mediates direct toxicity to nerve cells, and eventually simulates complex physiological processes such as activation of microglia to repair the damaged area. Herein, we show that sprouty-related protein with an EVH1 domain 1 (SPRED1) may act as a regulatory node in this phenomenon. The ischemic brain of an ischemia-reperfusion rat model constructed by middle cerebral artery occlusion (MCAO) showed an increase in oxidative stress and downregulation of SPRED1 expression. Hydrogen peroxide (H2O2)-simulated oxidative damage exerted a fluctuating regulatory effect on SPRED1 level in BV2 microglia, which is highly consistent with its regulatory effect on nuclear factor kappa B (NF-κB) transcription factor p65. Interestingly, SPRED1 overexpressed in BV2 cells did not exert any regulatory effect on p38 mitogen-activated protein kinase (MAPK), NF-κB p65, and pro-inflammatory cytokines. However, treatment of BV2 cells overexpressing SPRED1 with H2O2 led to significant changes in the above phenomena as well as their viability and apoptosis. In the absence of H2O2 induction, SPRED overexpression alone did not mediate such an effect. These findings indicate that SPRED1 tends to maintain intracellular homeostasis of signals, but the oxidative stress derived from ischemia-reperfusion can easily degrade SPRED1 and consequently re-activate these restricted signals and alter the behavior of microglia. Thus, our study reveals a novel role of SPRED1 in microglia in response to cerebral ischemia-induced oxidative stress.
Collapse
Affiliation(s)
- Li Wei
- Department of Blood Transfusion, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xin Li
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qianfeng Wei
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lin Chen
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Li Xu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| | - Peng Zhou
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
22
|
Xiang Y, Tan M, Ning Z, Zhang Y. Anti-cerebral ischemic neuronal injury mechanism of Zhenlong Xingnao capsules: role of the Notch/NF-κB signaling pathway. Am J Transl Res 2023; 15:4587-4599. [PMID: 37560215 PMCID: PMC10408513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/19/2023] [Indexed: 08/11/2023]
Abstract
OBJECTIVE To investigate the anti-cerebral ischemia-reperfusion injury (CIRI) effect and mechanism of Zhenlong Xingnao capsules based on Notch/NF-κB signaling pathway. METHODS The rat model of middle cerebral artery occlusion (MCAO) was established using the Longa suture occlusion method, and 70 rats were divided into sham-operated, model, low dose Zhenlong Xingnao capsule group (125 mg/kg Zhenlong Xingnao capsule solution) and high dose Zhenlong Xingnao capsule group (250 mg/kg Zhenlong Xingnao capsule solution), low dose Zhenlong Xingnao capsule + neurogenic site notch homologous protein 1 (Notch1) antibody (Jagged1 group, 125 mg/kg capsule solution + 25 mg/kg Jagged1 solution), high dose Zhenlong Xingnao capsule + Jagged1 group (250 mg/kg capsule solution + 25 mg/kg Jagged1 solution), and Jagged1 group (25 mg/kg Jagged1 solution). The learning and memory abilities (behavioral score, spontaneous movement, and rotarod test), neurological function score, inflammatory factors and oxidative stress levels [interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD)] in hippocampal tissue, and Bcl-2, Bax, and Caspase-3 mRNA levels were measured by reverse transcription quantitative polymerase chain reaction, and Notch1/NF-κB signaling pathway-related protein expression was assessed by Western blot. RESULTS The low and high dose interventions of Zhenlong Xingnao capsules significantly improved the learning and memory abilities of MCAO rats, reduced the neurological impairment scores, improved the levels of IL-6, TNF-α, MDA, GSH-Px, SOD, and inhibited the expression levels of Notch1, p-NF-κB p65, and Hes-1 proteins. However, the protective effect of Zhenlong Xingnao capsules on neurons in rat brain tissue could be reduced after treatment with Jagged1. CONCLUSIONS Zhenlong Xingnao capsules can promote neuronal repair during ischemia-reperfusion, and its mechanism may be related to inhibiting the activation of Notch/NF-κB signaling pathway and reducing inflammation and oxidative stress response.
Collapse
Affiliation(s)
- Yingqing Xiang
- School of Medicine, Jiangxi University of Technology Nanchang 330098, Jiangxi, China
| | - Meichun Tan
- School of Medicine, Jiangxi University of Technology Nanchang 330098, Jiangxi, China
| | - Zhen Ning
- School of Medicine, Jiangxi University of Technology Nanchang 330098, Jiangxi, China
| | - Yuhua Zhang
- School of Medicine, Jiangxi University of Technology Nanchang 330098, Jiangxi, China
| |
Collapse
|
23
|
Wei W, Li H, Deng Y, Zheng X, Zhou Y, Xue X. The combination of Alisma and Atractylodes ameliorates cerebral ischaemia/reperfusion injury by negatively regulating astrocyte-derived exosomal miR-200a-3p/141-3p by targeting SIRT1. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116597. [PMID: 37146842 DOI: 10.1016/j.jep.2023.116597] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/22/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The combination of Alisma and Atractylodes (AA), a classical traditional Chinese herbal decoction, may protect against cerebral ischaemia/reperfusion injury (CIRI). However, the underlying mechanism has not been characterized. Intriguingly, exosomal microRNAs (miRNAs) have been recognized as vital factors in the pharmacology of Chinese herbal decoctions. AIM OF THE STUDY The aim of the present study was to assess whether the neuroprotective effect of AA was dependent on the efficient transfer of miRNAs via exosomes in the brain. MATERIALS AND METHODS Bilateral common carotid artery ligation (BCAL) was used to induce transient global cerebral ischaemia/reperfusion (GCI/R) in C57BL/6 mice treated with/without AA. Neurological deficits were assessed with the modified neurological severity score (mNSS) and Morris water maze (MWM) test. Western blot (WB) analysis was used to detect the expression of sirtuin 1 (SIRT1) in the cerebral cortex. The inflammatory state was quantitatively evaluated by measuring the expression of phospho-Nuclear factor kappa B (p-NF-κB), Interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) using WB analysis and glial fibrillary acidic protein (GFAP) immunohistochemical staining. The protein expression of zonula occluden-1 (ZO-1), occludin, caudin-5 and CD31 was examined by immunohistochemical staining to determine blood‒brain barrier (BBB) permeability. Exosomes were extracted from the brain interstitial space by ultracentrifugation and identified by transmission electron microscopy (TEM), WB analysis and nanoparticle tracking analysis (NTA). The origin of exosomes was clarified by measuring the specific mRNAs within exosomes via Real Time Quantitative PCR (RT‒qPCR). Differential miRNAs in exosomes were identified using microarray screening and were validated by RT‒qPCR. Exosomes were labelled with fluorescent dye (PKH26) and incubated with bEnd.3 cells, the supernatant was collected, IL-1β/TNF-α expression was measured using enzyme-linked immunosorbent assay (ELISA), total RNA was extracted, and miR-200a-3p/141-3p expression was examined by RT‒qPCR. In addition, the levels of miR-200a-3p/141-3p in oxygen glucose deprivation/reoxygenation (OGD/R)-induced bEnd.3 cells were quantified. The direct interaction between miR-200a-3p/141-3p and the SIRT1 3' untranslated region (3'UTR) was measured by determining SIRT1 expression in bEnd.3 cells transfected with the miR-200a-3p/141-3p mimic/inhibitor. RESULTS Severe neurological deficits and memory loss caused by GCI/R in mice was markedly ameliorated by AA treatment, particularly in the AA medium-dose group. Moreover, AA-treated GCI/R-induced mice showed significant increases in SIRT1, ZO-1, occludin, caudin-5, and CD31 expression levels and decreases in p-NF-κB, IL-1β, TNF-α, and GFAP expression levels compared with those in untreated GCI/R-induced mice. Furthermore, we found that miR-200a-3p/141-3p was enriched in astrocyte-derived exosomes from GCI/R-induced mice and could be inhibited by treatment with a medium dose of AA. The exosomes mediated the transfer of miR-200a-3p/141-3p into bEnd.3 cells, promoted IL-1β and TNF-α release and downregulated the expression of SIRT1. No significant changes in the levels of miR-200a-3p/141-3p were observed in OGD/R-induced bEnd.3 cells. The miR-200a-3p/141-3p mimic/inhibitor decreased/increased SIRT1 expression in bEnd.3 cells, respectively. CONCLUSION Our findings demonstrated that AA attenuated inflammation-mediated CIRI by inhibiting astrocyte-derived exosomal miR-200a-3p/141-3p by targeting the SIRT1 gene, which provided further evidence and identified a novel regulatory mechanism for the neuroprotective effects of AA.
Collapse
Affiliation(s)
- Wei Wei
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huihong Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - YunFei Deng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - XiaoQing Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yangjie Zhou
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.
| | - Xiehua Xue
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Fujian Key Laboratory of Rehabilitation Technology, Fujian Key Laboratory of Cognitive Rehabilitation, Fuzhou, China.
| |
Collapse
|
24
|
Zhang W, Wu Y, Zeng M, Yang C, Qiu Z, Liu R, Wang L, Zhong M, Chen Q, Liang W. Protective role of remote ischemic conditioning in renal transplantation and partial nephrectomy: A systematic review and meta-analysis of randomized controlled trials. Front Surg 2023; 10:1024650. [PMID: 37091267 PMCID: PMC10113469 DOI: 10.3389/fsurg.2023.1024650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
Objective Studies have shown that remote ischemic conditioning (RIC) can effectively attenuate ischemic-reperfusion injury in the heart and brain, but the effect on ischemic-reperfusion injury in patients with kidney transplantation or partial nephrectomy remains controversial. The main objective of this systematic review and meta-analysis was to investigate whether RIC provides renal protection after renal ischemia-reperfusion injury in patients undergoing kidney transplantation or partial nephrectomy. Methods A computer-based search was conducted to retrieve relevant publications from the PubMed database, Embase database, Cochrane Library and Web of Science database. We then conducted a systematic review and meta-analysis of randomized controlled trials that met our study inclusion criteria. Results Eleven eligible studies included a total of 1,145 patients with kidney transplantation or partial nephrectomy for systematic review and meta-analysis, among whom 576 patients were randomly assigned to the RIC group and the remaining 569 to the control group. The 3-month estimated glomerular filtration rate (eGFR) was improved in the RIC group, which was statistically significant between the two groups on kidney transplantation [P < 0.001; mean difference (MD) = 2.74, confidence interval (CI): 1.41 to 4.06; I 2 = 14%], and the 1- and 2-day postoperative Scr levels in the RIC group decreased, which was statistically significant between the two groups on kidney transplantation (1-day postoperative: P < 0.001; MD = 0.10, CI: 0.05 to 0.15, I 2 = 0; 2-day postoperative: P = 0.006; MD = 0.41, CI: 0.12 to 0.70, I 2 = 0), but at other times, there was no significant difference between the two groups in Scr levels. The incidence of delayed graft function (DGF) decreased, but there was no significant difference (P = 0.60; 95% CI: 0.67 to 1.26). There was no significant difference between the two groups in terms of cross-clamp time, cold ischemia time, warm ischemic time, acute rejection (AR), graft loss or length of hospital stay. Conclusion Our meta-analysis showed that the effect of remote ischemia conditioning on reducing serum creatinine (Scr) and improving estimate glomerular filtration rate (eGFR) seemed to be very weak, and we did not observe a significant protective effect of RIC on renal ischemic-reperfusion. Due to small sample sizes, more studies using stricter inclusion criteria are needed to elucidate the nephroprotective effect of RIC in renal surgery in the future.
Collapse
Affiliation(s)
- Wenfu Zhang
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
- Department of Anesthesia, hospital of Traditional Chinese Medicine of Zhongshan, Zhongshan, China
| | - Yingting Wu
- Department of Critical Care Medicine Nursing, the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mingwang Zeng
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Chao Yang
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Zhengang Qiu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Rongrong Liu
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Lifeng Wang
- Anesthesia Surgery Center of the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Maolin Zhong
- Anesthesia Surgery Center of the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qiaoling Chen
- Department of Anesthesiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Weidong Liang
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
- Anesthesia Surgery Center of the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
25
|
Wang T, Xie ZH, Wang L, Luo H, Zhang J, Dong WT, Zheng XH, Ye C, Tian XB, Liu G, Zhu XS, Li YL, Kang QL, Zhang F, Peng WX. LncAABR07053481 inhibits bone marrow mesenchymal stem cell apoptosis and promotes repair following steroid-induced avascular necrosis. Commun Biol 2023; 6:365. [PMID: 37012358 PMCID: PMC10070412 DOI: 10.1038/s42003-023-04661-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 03/03/2023] [Indexed: 04/05/2023] Open
Abstract
The osteonecrotic area of steroid-induced avascular necrosis of the femoral head (SANFH) is a hypoxic microenvironment that leads to apoptosis of transplanted bone marrow mesenchymal stem cells (BMSCs). However, the underlying mechanism remains unclear. Here, we explore the mechanism of hypoxic-induced apoptosis of BMSCs, and use the mechanism to improve the transplantation efficacy of BMSCs. Our results show that the long non-coding RNA AABR07053481 (LncAABR07053481) is downregulated in BMSCs and closely related to the degree of hypoxia. Overexpression of LncAABR07053481 could increase the survival rate of BMSCs. Further exploration of the downstream target gene indicates that LncAABR07053481 acts as a molecular "sponge" of miR-664-2-5p to relieve the silencing effect of miR-664-2-5p on the target gene Notch1. Importantly, the survival rate of BMSCs overexpressing LncAABR07053481 is significantly improved after transplantation, and the repair effect of BMSCs in the osteonecrotic area is also improved. This study reveal the mechanism by which LncAABR07053481 inhibits hypoxia-induced apoptosis of BMSCs by regulating the miR-664-2-5p/Notch1 pathway and its therapeutic effect on SANFH.
Collapse
Affiliation(s)
- Tao Wang
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Zhi-Hong Xie
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Lei Wang
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Hong Luo
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Jian Zhang
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Wen-Tao Dong
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Xiao-Han Zheng
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Chuan Ye
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Xiao-Bin Tian
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Gang Liu
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China
| | - Xue-Song Zhu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, P.R. China
| | - Yan-Lin Li
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, P.R. China
| | - Qing-Lin Kang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, P.R. China
| | - Fei Zhang
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China.
| | - Wu-Xun Peng
- Department of Orthopedics and Traumatology, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, P.R. China.
| |
Collapse
|
26
|
Du Y, Qiu R, Chen L, Chen Y, Zhong Z, Li P, Fan F, Cheng Y. Identification of serum exosomal metabolomic and proteomic profiles for remote ischemic preconditioning. J Transl Med 2023; 21:241. [PMID: 37009888 PMCID: PMC10069038 DOI: 10.1186/s12967-023-04070-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/18/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Remote ischemic preconditioning (RIPC) refers to a brief episode of exposure to potential adverse stimulation and prevents injury during subsequent exposure. RIPC has been shown to increase tolerance to ischemic injury and improve cerebral perfusion status. Exosomes have a variety of activities, such as remodeling the extracellular matrix and transmitting signals to other cells. This study aimed to investigate the potential molecular mechanism of RIPC-mediated neuroprotection. METHODS Sixty adult male military personnel participants were divided into the control group (n = 30) and the RIPC group (n = 30). We analyzed the differential metabolites and proteins in the serum exosomes of RIPC participants and control subjects. RESULTS Eighty-seven differentially expressed serum exosomal metabolites were found between the RIPC and control groups, which were enriched in pathways related to tyrosine metabolism, sphingolipid metabolism, serotonergic synapses, and multiple neurodegeneration diseases. In addition, there were 75 differentially expressed exosomal proteins between RIPC participants and controls, which involved the regulation of insulin-like growth factor (IGF) transport, neutrophil degranulation, vesicle-mediated transport, etc. Furthermore, we found differentially expressed theobromine, cyclo gly-pro, hemopexin (HPX), and apolipoprotein A1 (ApoA1), which are associated with neuroprotective benefits in ischemia/reperfusion injury. In addition, five potential metabolite biomarkers, including ethyl salicylate, ethionamide, piperic acid, 2, 6-di-tert-butyl-4-hydroxymethylphenol and zerumbone, that separated RIPC from control individuals were identified. CONCLUSION Our data suggest that serum exosomal metabolites are promising biomarkers for RIPC, and our results provide a rich dataset and framework for future analyses of cerebral ischemia‒reperfusion injury under ischemia/reperfusion conditions.
Collapse
Affiliation(s)
- Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Rui Qiu
- Institute of National Security, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifeng Zhong
- Department of High Altitude Operational Medicine, College of High Altitude Military Medicine, Army Medical University, (Third Military Medical University), Chongqing, China
| | - Peng Li
- Department of High Altitude Operational Medicine, College of High Altitude Military Medicine, Army Medical University, (Third Military Medical University), Chongqing, China
| | - Fangcheng Fan
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China.
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China.
- Institute of National Security, Minzu University of China, Beijing, China.
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
| |
Collapse
|
27
|
Lu H, Chen S, Nie Q, Xue Q, Fan H, Wang Y, Fan S, Zhu J, Shen H, Li H, Fang Q, Ni J, Chen G. Synaptotagmin-3 interactions with GluA2 mediate brain damage and impair functional recovery in stroke. Cell Rep 2023; 42:112233. [PMID: 36892998 DOI: 10.1016/j.celrep.2023.112233] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 01/20/2023] [Accepted: 02/23/2023] [Indexed: 03/10/2023] Open
Abstract
Synaptotagmin III (Syt3) is a Ca2+-dependent membrane-traffic protein that is highly concentrated in synaptic plasma membranes and affects synaptic plasticity by regulating post-synaptic receptor endocytosis. Here, we show that Syt3 is upregulated in the penumbra after ischemia/reperfusion (I/R) injury. Knockdown of Syt3 protects against I/R injury, promotes recovery of motor function, and inhibits cognitive decline. Overexpression of Syt3 exerts the opposite effects. Mechanistically, I/R injury augments Syt3-GluA2 interactions, decreases GluA2 surface expression, and promotes the formation of Ca2+-permeable AMPA receptors (CP-AMPARs). Using a CP-AMPAR antagonist or dissociating the Syt3-GluA2 complex via TAT-GluA2-3Y peptide promotes recovery from neurological impairments and improves cognitive function. Furthermore, Syt3 knockout mice are resistant to cerebral ischemia because they show high-level expression of surface GluA2 and low-level expression of CP-AMPARs after I/R. Our results indicate that Syt3-GluA2 interactions, which regulate the formation of CP-AMPARs, may be a therapeutic target for ischemic insults.
Collapse
Affiliation(s)
- Haifeng Lu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Shujun Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Qianqian Nie
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Qun Xue
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Yiqing Wang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Shenghao Fan
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Juehua Zhu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Jianqiang Ni
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Institute of Stroke Research, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
28
|
Yang X, Xu L, Zhao H, Xie T, Wang J, Wang L, Yang J. Curcumin protects against cerebral ischemia-reperfusion injury in rats by attenuating oxidative stress and inflammation: a meta-analysis and mechanism exploration. Nutr Res 2023; 113:14-28. [PMID: 36996692 DOI: 10.1016/j.nutres.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/11/2023] [Accepted: 02/26/2023] [Indexed: 03/09/2023]
Abstract
Accumulating evidence has suggested that curcumin may protect against cerebral ischemia-reperfusion injury (CIRI). However, biological mechanisms vary across studies, limiting the clinical applicability of these findings. We performed a meta-analysis on publications evaluating curcumin administration in rat models of CIRI. Furthermore, we sought to test the hypothesis that curcumin alleviates CIRI through diminishing oxidation and inflammation. We searched PubMed, Embase, Web of Science, and Cochrane from the starting date of each database to May 2022 for experimental rat studies exploring the use of curcumin after ischemia reperfusion. Included articles were assessed for bias using SYRCLE's risk of bias tool. Data were aggregated by a random effects model. Curcumin administration significantly reduced neurological deficit score (20 studies; pooled mean difference [MD] = -1.57; 95% CI, -1.78 to -1.36, P < .00001), infarct volume (18 studies; pooled MD = -17.56%; 95% CI, -20.92% to -14.20%; P < 0.00001), and brain water content (8 studies, pooled MD = -11.29%, 95% CI: -16.48%, -6.11%, P < .00001). Compared with control, the levels of superoxide dismutase, glutathione, and glutathione peroxidase were significantly higher, whereas the levels of reactive oxygen species, malondialdehyde, interleukin-1β, interleukin-6, interleukin-8, and nuclear factor kappa B were significantly lower (P < .05). Subgroup analysis raised the possibility that intervention affections differed by curcumin's dose. To our knowledge, this is the first meta-analysis of curcumin's neuroprotection and mechanisms in rat CIRI models. Our analysis suggests the neuroprotective potential of curcumin in CIRI via antioxidant activity and anti-inflammatory effect. More research is required to further confirm the effectiveness and safety of curcumin on ischemic stroke therapy.
Collapse
Affiliation(s)
- Xuyi Yang
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Liang Xu
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Hui Zhao
- Department of Critical Care Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Tinghui Xie
- School of Agriculture and Bioengineering, Taizhou Vocational College of Science and Technology, Taizhou, China
| | - Jiabing Wang
- Department of Pharmacy, Taizhou Municipal Hospital, Taizhou, China
| | - Lei Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianwei Yang
- General Practice, Zhejiang Taizhou Hospital, Linhai, China.
| |
Collapse
|
29
|
FTO inhibits oxidative stress by mediating m6A demethylation of Nrf2 to alleviate cerebral ischemia/reperfusion injury. J Physiol Biochem 2023; 79:133-146. [PMID: 36327034 DOI: 10.1007/s13105-022-00929-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Current therapies are of limited efficacy in cerebral ischemia/reperfusion (I/R) injury. Based on the important role of oxidative stress in cerebral I/R injury, this study aimed to explore how the N6-adenosine methylation (m6A) demethylase FTO affects oxidative stress. Middle cerebral artery occlusion/reperfusion (MCAO/R)-induced rat model and oxygen and glucose deprivation/re-oxygenation (OGD/R)-induced SH-SY5Y cells were established as in vivo and in vitro model, respectively. The neurological score of rats was measured, and the volume of cerebral infarction was measured by TTC staining. The levels of FTO, nuclear factor-erythroid 2-related factor (Nrf2), and the activity of m6A demethylase FTO were detected. The m6A methylation level of Nrf2 mRNA was detected by MeRIP experiment. Flow cytometry and MTT assay were used to detect apoptosis and proliferation in vitro. TUNEL assay was used to detect apoptosis in brain tissues. FTO and Nrf2 expressions were decreased in the MCAO/R rat brain tissues and OGD/R SH-SY5Y cells, while the m6A methylation level of Nrf2 mRNA was significantly increased. Overexpression of FTO upregulated Nrf2 expression by decreasing the m6A methylation level of Nrf2 mRNA. m6A binding protein YT521-B homology (YTH) domain family protein 2 (YTHDF2) promoted the degradation of Nrf2 by promoting the m6A methylation level of Nrf2 mRNA. Furthermore, SH-SY5Y cell apoptosis was increased and cell viability was decreased after the addition of methyltransferases METTL 3/14, thus blocking FTO to protect SH-SY5Y cells from oxidative stress injury. In vivo, overexpression of FTO decreased the area of cerebral ischemia infarction and the extent of cell apoptosis. In conclusion, FTO increases Nrf2 expression by mediating m6A demethylation of Nrf2 mRNA, thereby inhibiting oxidative stress response and ultimately alleviating cerebral I/R injury.
Collapse
|
30
|
Aldehyde Dehydrogenase 2 Protects the Kidney from Ischemia-Reperfusion Injury by Suppressing the I κB α/NF- κB/IL-17C Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2264030. [PMID: 36865346 PMCID: PMC9974261 DOI: 10.1155/2023/2264030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 02/25/2023]
Abstract
Objective Ischemia-reperfusion injury (IRI) is an important cause of delayed functional recovery after transplantation. This study is aimed at investigating the molecular mechanism of ALDH2 in a kidney ischemia-reperfusion model based on RNA-seq. Methods We performed kidney ischemia-reperfusion in ALDH2-/- and WT mice and evaluated kidney function and morphology using SCr, HE staining, TUNEL staining, and TEM. We used RNA-seq to compare mRNA expression in ALDH2-/- and WT mice after IR, and then, we verified the related molecular pathways by PCR and western blotting. In addition, activators and inhibitors of ALDH2 were used to alter the activity of ALDH2. Finally, we established a model of hypoxia and reoxygenation in HK-2 cells and clarified the role of ALDH2 in IR by interfering with ALDH2 and using an NF-κB inhibitor. Results After kidney ischemia-reperfusion, the SCr value increased significantly, kidney tubular epithelial cells were damaged, and the apoptosis rate increased. In the microstructure, mitochondria were swollen and deformed, and ALDH2 deficiency aggravated these changes. The NF-κB pathway and IL-17 pathway were significantly enriched in ALDH2-/- mice compared with WT mice according to KEGG enrichment analysis of the RNA-seq data. The PCR results showed that the mRNA expression levels of IκBα and IL-17B, C, D, E, and F were significantly higher than those in the WT-IR group. Western blot verification results showed that ALHD2 knockdown resulted in increased phosphorylation of IκBα, increased phosphorylation of NF-κB, and increased expression of IL-17C. When we used ALDH2 agonists, the number of lesions and the expression levels of the corresponding proteins were reduced. Knockdown of ALDH2 in HK-2 cells resulted in a higher proportion of apoptotic cells after hypoxia and reoxygenation, but inhibiting the phosphorylation of NF-κB prevented the increase in apoptosis and reduced the protein expression level of IL-17C. Conclusion ALDH2 deficiency can lead to the aggravation of kidney ischemia-reperfusion injury. RNA-seq analysis and validation by PCR and western blotting revealed that this effect may be due to the promotion of IκBα/NF-κB p65 phosphorylation during ischemia-reperfusion caused by ALDH2 deficiency, which then leads to an increase in inflammatory factors, including IL-17C. Thus, cell death is promoted, and kidney IRI is eventually aggravated. We link ALDH2 deficiency with inflammation, revealing a new idea for ALDH2-related research.
Collapse
|
31
|
Zhou Y, He Y, Yan S, Chen L, Zhang R, Xu J, Hu H, Liebeskind DS, Lou M. Reperfusion Injury Is Associated With Poor Outcome in Patients With Recanalization After Thrombectomy. Stroke 2023; 54:96-104. [PMID: 36367100 DOI: 10.1161/strokeaha.122.039337] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The existence of cerebral reperfusion injury in human beings remains controversial. Thus, we aimed to explore the presence of reperfusion injury in acute ischemic stroke patients with recanalization after mechanical thrombectomy and analyzed its impact on neurological outcome. METHODS We reviewed our prospectively collected database CIPPIS (Comparison Influence to Prognosis of CTP and MRP in AIS Patients, NCT03367286), and enrolled anterior circulation large artery occlusion patients with recanalization after mechanical thrombectomy who underwent (1) computed tomography (CT) perfusion on admission and immediately after recanalization to determine reperfusion region, and (2) CT and/or magnetic resonance imaging (MRI) immediately and 24 hours after recanalization to determine lesion areas. The expansion of lesion between recanalization and 24 hours within reperfusion region was potentially caused by reperfusion, thus termed as radiological observed reperfusion injury (RORI). Based on the imaging modality immediately after recanalization, RORI was further divided into RORICT and RORIMRI. We first included a small cohort who had performed both CT and MRI immediately after recanalization to validate the consistency between RORICT and RORIMRI (Study 1). Then the association with RORICT and poor outcome, defined as 3-month modified Rankin Scale score of 3 to 6, was explored in a larger cohort (Study 2). RESULTS Study 1 included 23 patients and good consistency was found between RORICT and RORIMRI (intraclass correlation=0.97, P<0.001). Among 226 patients included in Study 2, a total of 106 (46.9%) were identified with RORI. The ratio of RORI to reperfusion region was 30.1 (16.2, 51.0)% and was independently associated with poor outcome (odds ratio=1.55 per 10% [95% CI' 1.30-1.84]; P<0.001). CONCLUSIONS Our findings suggested that RORI was relatively frequent in stroke patients with recanalization after mechanical thrombectomy and associated with poor outcome despite successful recanalization. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03367286.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Yaode He
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Shenqiang Yan
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Lin Chen
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Ruoxia Zhang
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Jinjin Xu
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | - Haitao Hu
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| | | | - Min Lou
- Department of Neurology, the 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (Y.Z., Y.H., S.Y., L.C., R.Z., J.X., H.H., M.L.)
| |
Collapse
|
32
|
Li L, Pan G, Fan R, Li D, Guo L, Ma L, Liang H, Qiu J. Luteolin alleviates inflammation and autophagy of hippocampus induced by cerebral ischemia/reperfusion by activating PPAR gamma in rats. BMC Complement Med Ther 2022; 22:176. [PMID: 35778706 PMCID: PMC9248165 DOI: 10.1186/s12906-022-03652-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background Luteolin, a flavonoid compound with anti-inflammatory activity, has been reported to alleviate cerebral ischemia/reperfusion (I/R) injury. However, its potential mechanism remains unclear. Methods The binding activity of luteolin to peroxisome proliferator-activated receptor gamma (PPARγ) was calculated via molecular docking analysis. Rats were subjected to middle cerebral artery occlusion and reperfusion (MCAO/R). After reperfusion, vehicle, 25 mg/kg/d luteolin, 50 mg/kg/d luteolin, 10 mg/kg/d pioglitazone, 50 mg/kg/d luteolin combined with 10 mg/kg/d T0070907 (PPARγ inhibitor) were immediately orally treatment for 7 days. ELISA, TTC staining, H&E staining, immunohistochemistry, immunofluorescence and transmission electron microscope methods were performed to evaluate the inflammation and autophagy in damaged hippocampal region. The PPARγ, light chain 3 (LC3) B-II/LC3B-I and p-nuclear factor-κB (NF-κB) p65 proteins expression levels in damaged hippocampal region were analyzed. Results Luteolin showed good PPARγ activity according to docking score (score = − 8.2). Luteolin treatment downregulated the infarct area and the pro-inflammatory cytokines levels caused by MCAO/R injury. Moreover, luteolin administration ameliorated neuroinflammation and autophagy in damaged hippocampal region. Pioglitazone plays protective roles similar to luteolin. T0070907 concealed the neuroprotective roles of 50 mg/kg/d luteolin. Conclusions Luteolin exerts neuroprotective roles against inflammation and autophagy of hippocampus induced by cerebral I/R by activating PPARγ in rats. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03652-8.
Collapse
|
33
|
Xue LX, Chen SF, Xue SX, Liu PD, Liu HB. LncRNA TUG1 compromised neuronal mitophagy in cerebral ischemia/reperfusion injury by targeting sirtuin 1. Cell Biol Toxicol 2022; 38:1121-1136. [PMID: 35348966 DOI: 10.1007/s10565-022-09700-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 02/11/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Mitophagy protects against cerebral ischemia/reperfusion (CI/R)-induced neuronal apoptosis via mitochondrial clearance. Although taurine-upregulated gene 1 (lncRNA TUG1) has been proposed to be involved in the neuronal apoptosis evoked by CI/R, its specific role in mitophagy during the progression of CI/R injury remains unknown. METHODS The CI/R rat model was established using middle cerebral artery occlusion/reperfusion (MCAO/R). Human neuroblastoma cell line SH-SY5Y was subjected to oxygen-glucose deprivation and reoxygenation (OGD/R). Ubiquitination assay, co-immunoprecipitation assay, RNA pull-down, and RNA immunoprecipitation were used to determine the interplay among TUG1, sirtuin 1 (SIRT1), and F-box and WD repeat domain-containing 7 (FBXW7). RESULTS The upregulation of the TUG1 level and downregulation of the mitophagy were observed in both MCAO/R-treated rats and OGD/R-treated cells. The administration of si-TUG1 (a siRNA directed against TUG1) potentiated mitophagy and suppressed neuronal apoptosis in OGD/R-treated cells. However, the neuroprotective effect of si-TUG1 was reversed by mitophagy inhibitor or SIRT1 knockdown in vitro. Functionally, TUG1 enhanced FBXW7-mediated SIRT1 ubiquitination by upregulating FBXW7 expression. The overexpression of FBXW7 abrogated the si-TUG1-reinforced mitophagy by decreasing SIRT1 expression, thus aggravating neuronal apoptosis in the OGD/R+si-TUG1-treated cells. In rats with MCAO/R, the interference of TUG1 clearly decreased neuronal apoptosis, lessened the infarct volume, and relieved the neurological deficits. CONCLUSION TUG1 knockdown promotes SIRT1-induced mitophagy by suppressing FBXW7-mediated SIRT1 degradation, thus relieving the neuronal apoptosis induced by CI/R injury. LncRNA TUG1 promotes neuronal apoptosis through inhibition of mitophagy. TUG1 decreased SIRT1 expression by promoting FBXW7-mediated SIRT1 ubiquitination. FBXW7/SIRT1 axis mediated the effect of TUG1 on OGD/R-induced neuronal apoptosis by regulating mitophagy.
Collapse
Affiliation(s)
- Long-Xing Xue
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 JianShe Road, Zhengzhou, 450052, Henan Province, People's Republic of China
| | - Song-Feng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shi-Xing Xue
- Department of Neurology, Affiliated Xinhua Hospital of Dalian University, Dalian, 116021, China
| | - Pei-Dong Liu
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Hong-Bo Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 JianShe Road, Zhengzhou, 450052, Henan Province, People's Republic of China.
| |
Collapse
|
34
|
Xu Y, Deng T, Xie L, Qin T, Sun T. Neuroprotective effects of hawthorn leaf flavonoids in
Aβ
25–35
‐induced
Alzheimer's disease model. Phytother Res 2022; 37:1346-1365. [PMID: 36447359 DOI: 10.1002/ptr.7690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/17/2022] [Accepted: 11/06/2022] [Indexed: 12/02/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by β-amyloid (Aβ) plaques, neurofibrillary tangles, neuronal cell loss, and oxidative stress. Further deposition of Aβ in the brain induces oxidative stress, neuroinflammation, and memory dysfunction. Hawthorn (Crataegus pinnatifida Bge.) leaf, a known traditional Chinese medicine, is commonly used for the treatment of hyperlipidemia, heart palpitations, forgetfulness, and tinnitus, and its main bioactive components are Hawthorn Leaf Flavonoids (HLF). In this study, we investigated the neuroprotective effects of the HLF on the Aβ25-35 (bilateral hippocampus injection) rat model of AD. The results showed that the oral administration of HLF at a dose of 50, 100, and 200 mg/kg for 30 days significantly ameliorated neuronal cell damage and memory deficits, and markedly increased the enzyme activities of superoxide dismutase and catalase, and the content of glutathione whereas it decreased the malondialdehyde content in the Aβ25-35 rat model of AD as well as suppressed the activation of astrocytes. In addition, HLF up-regulated Nrf-2, NQO-1, and HO-1 protein expressions. Also, it reduced neuroinflammation by inhibiting activation of astrocytes. In summary, these results indicated that HLF decreased the oxidative stress via activating Nrf-2/antioxidant response element signaling pathways, and may suggest as a potential candidate for AD therapeutic agent.
Collapse
Affiliation(s)
- Ying Xu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine Chengdu People's Republic of China
| | - Ting Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu People's Republic of China
| | - Linjiang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu People's Republic of China
| | - Tao Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu People's Republic of China
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu People's Republic of China
| |
Collapse
|
35
|
Lu Y, Pang Q, Wu Q, Luo B, Tang X, Jiang Q. Molar loss further exacerbates 2-VO-induced cognitive impairment associated with the activation of p38MAPK/NFκB pathway. Front Aging Neurosci 2022; 14:930016. [DOI: 10.3389/fnagi.2022.930016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
BackgroundVascular dementia is characterized by reduced cognitive function due to chronic cerebral hypoperfusion and has become a significant public health challenge as the global population ages. Recent studies suggested that molar loss, a common problem among the elderly, may trigger the development of cognitive decline. Our previous study found that the molar loss affected cognitive dysfunction, and the astrocytes in the hippocampus of chronic cerebral ischemia rats were affected, but the underlying mechanism is unclear.MethodsIn this study, we established the animal model of molar loss with 2-VO rats and the Morris water maze was used to test the cognitive ability of rats in each group. The damage to neurons was observed via Nissl staining, and neuronal apoptosis was analyzed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay in the hippocampus of the rats. Quantitative Real-Time PCR and immunohistochemistry and histology (IHC) were used to detect the expression of p38MAPK, NFκB, caspase 3, and iNOS in the hippocampus. The astrocytes were detected by IHC and Immunofluorescence analysis for GFAP. After 2-VO MO surgery, rats were administered DMSO or p38MAPK inhibitor (SB203580) by intrathecal injection.ResultsThe Morris water maze test showed that the molar loss aggravated spatial memory learning ability with chronic cerebral ischemia decreased in the rats. The neuronal damage and more apoptotic cells were observed in the hippocampus of 2-VO rats. After the molar loss, the mRNA and protein expression of iNOS, p38MAPK, NFκB, and caspase 3 were further upregulated in 2-VO rats. Molar loss upregulated GFAP expression, and the p38MAPK-positive cells were labeled with the astrocyte marker GFAP. SB203580 reduced cognitive impairment and apoptosis of hippocampal neurons in 2-VO rats following the molar loss.ConclusionMolar loss can aggravate cognitive impairment in 2-VO rats to a certain extent. The mechanism of molar loss exacerbating the cognitive decline in 2-VO rats may be associated with the activation of the p38MAPK-NFκB-caspase 3 signaling pathway, which induces neuronal apoptosis.
Collapse
|
36
|
Zhang L, Sui S, Wang S, Sun J. Neuroprotective Effect of Corosolic Acid Against Cerebral Ischemia-Reperfusion Injury in Experimental Rats. J Oleo Sci 2022; 71:1501-1510. [PMID: 36089398 DOI: 10.5650/jos.ess22130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Several therapeutic approaches were also urgently needed as ischaemic stroke was one of the most common brain disorders. Many phytochemicals have recently been discovered for the advancement of lead-like libraries that are concentrated on the peripheral and central nervous systems. Science does not yet understand how these drugs work, nor do they comprehend their in vivo characteristics. We investigated the potential benefits of corosolic acid (CA) in the treatment of brain injury caused by ischemia/reperfusion (I/R) in adult male Sprague-Dawley rats. Injury occurs after a 2-hour transient occlusion of the posterior cerebral artery and subsequent reperfusion (after 20 hours). Furthermore, the experiment assessed the size of the infarct, the amount of brain water present, as well as the neurofunctional conditions in rats. In the study, several markers of inflammation and cytokines associated with brain injury were measured. The Elisa kit was used in this study to measure the mRNA expression of interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin 1β, TNF-α (tumor necrosis factor), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and nitrous oxide (NO). The CA treatment significantly reduced brain water content, brain infarction volume, neurological scores, and Evans blue leakage (p < 0.001 and p < 0.001). Experimental rats were treated with CA after a significantly reduced level of anti-inflammatory, pro-inflammatory, and oxidative stress mediators was noted in their body tissues and serum (p < 0.001). By suppressing inflammatory responses in rats, CA demonstrated anti-inflammatory and neuroprotective properties.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurology, Central Hospital Affiliated to Shandong First Medical University
| | - Songtao Sui
- Department of Neurosurgery, Qingdao West Coast New Area Central Hospital
| | - Si Wang
- Department of Neurology, Central Hospital Affiliated to Shandong First Medical University
| | - Jinbo Sun
- Department of Neurology, Central Hospital Affiliated to Shandong First Medical University
| |
Collapse
|
37
|
Wang T, Xue Y, Li Y, Gao S, Peng L, Zhao Y, Yu S. DJ-1 Protein Inhibits Apoptosis in Cerebral Ischemia by Regulating the Notch1 and Nuclear Factor Erythroid2-Related Factor 2 Signaling Pathways. Neuroscience 2022; 504:33-46. [PMID: 36167256 DOI: 10.1016/j.neuroscience.2022.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022]
Abstract
DJ-1 plays a neuroprotective role in cerebral ischemia- reperfusion (I/R) injury and participates in the apoptosis of brain nerve cells, but the underlying mechanism is unclear. We explored the molecular pathways underlying this role using in vivo and in vitro approaches. Middle cerebral artery occlusion- reperfusion (MCAO/R) rat models and oxygen- glucose deprivation- reoxygenation (OGD/R) HAPI cell cultures were used to simulate cerebral ischemia-reperfusion injury. The interaction between DJ-1 and Notch1 was enhanced after MCAO/R in rats. After treatment of rats with DJ-1 siRNA, the expression of Notch1 and Nrf2 was down-regulated, and apoptosis was promoted. In contrast, the DJ-1 based peptide ND-13 upregulated the expression of Notch1 and Nrf2, and prevented apoptosis. In vitro, the Notch1 signaling pathway inhibitor DAPT reversed the neuroprotective effect of ND-13 and promoted apoptosis, weakened the interaction between DJ-1 and Notch1, and decreased the expression of proteins in the Notch1 and Nrf2 pathways. Thus, we found that DJ-1 inhibits apoptosis by regulating the Notch1 signaling pathway and Nrf2 expression in cerebral I/R injury. These results imply that DJ-1 is a potential therapeutic target for cerebral I/R injury.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China; Department of Pathology, Lu'an Hospital of Anhui Medical University, 237000 Anhui, People's Republic of China
| | - Ying Xue
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Yumei Li
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Sihao Gao
- Children's Hospital, Chongqing Medical University, 400014 Chongqing, People's Republic of China
| | - Li Peng
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.
| |
Collapse
|
38
|
The Neuroprotection Effects of Exosome in Central Nervous System Injuries: a New Target for Therapeutic Intervention. Mol Neurobiol 2022; 59:7152-7169. [DOI: 10.1007/s12035-022-03028-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 09/05/2022] [Indexed: 11/25/2022]
|
39
|
Wang L, Lai S, Zou H, Zhou X, Wan Q, Luo Y, Wu Q, Wan L, Liu J, Huang H. Ischemic preconditioning/ischemic postconditioning alleviates anoxia/reoxygenation injury via the Notch1/Hes1/VDAC1 axis. J Biochem Mol Toxicol 2022; 36:e23199. [PMID: 35975741 DOI: 10.1002/jbt.23199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/06/2022] [Accepted: 08/05/2022] [Indexed: 11/07/2022]
Abstract
Ischemic preconditioning (IPC), and ischemic postconditioning (IPost) have a significant protective effect on myocardial ischemia/reperfusion (MI/R) injury by alleviating oxidative stress and mitochondrial disturbances, although the underlying molecular mechanisms are unclear. The study was to demonstrate that cardioprotection against anoxia/reoxygenation (A/R) injury is transduced via the Notch1/Hes1/VDAC1 signaling pathway. Using mass spectrometry and tandem affinity purification (TAP), to screen for differentially expressed proteins associated with Hes1, followed by standard bioinformatics analysis. The co-immunoprecipitation (Co-IP) assay confirmed an interaction between Hes1 and VDAC1 proteins. H9c2 cells were transfected with Hes1 adenoviral N-terminal TAP vector (AD-NTAP/Hes1) and Hes1-short hairpin RNA adenoviral vector (AD-Hes1-shRNA) to establish A/R injury, IPC, and IPost models, respectively. The expression of Hes1 and VDAC1 proteins were measured by western blot analysis, while the levels of reactive oxygen species (ROS), mitochondrial membrane potential (ΔΨm), and apoptosis were evaluated by flow cytometry. AD-NTAP/Hes1 can activate the exogenous protein expression of Hes1, thus decreasing creatine phosphokinase (CPK) and lactate dehydrogenase (LDH) activity and promoting cell viability. The study found that VDAC1 was a potential target protein for Hes1 and the overexpression of Hes1 protein expression downregulated protein expression levels of VDAC1, reduced ROS production, stabilized ΔΨm, and inhibited apoptosis in H9c2 cells. Additionally, downregulation of Hes1 protein expression also upregulated VDAC1 protein expression, increased ROS production, imbalanced ΔΨm, promoted cell apoptosis, and attenuated the cardioprotection afforded by IPC and IPost. The Notch1/Hes1 signaling pathway activated by IPC/IPost can directly downregulate the protein expression of VDAC1 and consequently relieve A/R injury.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | | | - Huaxi Zou
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xueliang Zhou
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qing Wan
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yong Luo
- Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Qicai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Li Wan
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jichun Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Department of Cardiac Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Huang Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
40
|
Wang Q, Ma X. Gut microbial sodium butyrate alleviates renal ischemia-reperfusion injury by regulating HES1/PPARα. Mol Immunol 2022; 150:20-28. [PMID: 35930845 DOI: 10.1016/j.molimm.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022]
Abstract
This study investigated the effect of gut microbial sodium butyrate (NaB) on renal ischemia-reperfusion injury (IRI) and its mechanism using a rat model of renal IRI and a HK-2 cell model of hypoxia-reoxygenation (HR) injury. The activity of malondialdehyde, superoxide dismutase, glutathione peroxidase, and catalase in kidney tissues and HK-2 cells was detected. ELISA was performed to measure the concentrations of TNF-α, IL-1β, and IL-6 in serum and cell culture supernatant. TUNEL staining and flow cytometry were used to assess apoptosis in kidney tissues and HK-2 cells, respectively. UCSC and JASPAR predicted the binding sites between HES1 and PPARα promoter, followed by experimental verification of the binding. NaB pretreatment inhibited oxidative stress, inflammation, and apoptosis following renal IRI in vivo and in vitro. NaB suppressed the expression of HES1 and promoted that of PPARα. Overexpression of HES1 or knockdown of PPARα in HR-treated HK-2 cells inhibited the protective effects of NaB. HES1 repressed the expression of PPARα by binding PPARα promoter. In conclusion, NaB may alleviate renal IRI by promoting the transcription of PPARα via downregulation of HES1.
Collapse
Affiliation(s)
- Qiong Wang
- Kidney Disease and Dialysis Center, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, PR China
| | - Xiaoying Ma
- Kidney Disease and Dialysis Center, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, PR China; Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, PR China.
| |
Collapse
|
41
|
Zhang G, Guo J, Zeng J, Zhang X, Chen R, Wang G, Liang W. LncRNA SNHG14 is beneficial to oxygen glucose deprivation/reoxygenation-induced neuro-2a cell injury via mir-98-5p sequestration-caused BCL2L13 upregulation. Metab Brain Dis 2022; 37:2005-2016. [PMID: 35678981 DOI: 10.1007/s11011-022-01009-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The deregulation of long non-coding RNA (lncRNA) is associated with diverse human disorders, including cerebral ischemia/reperfusion injury (CI/RI). LncRNA SNHG14 was reported to function in CI/RI. Whereas, molecular mechanisms regulated by SNHG14 are not fully unveiled. METHODS Mice subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) were used as CI/RI animal models. Neuro-2a (N2A) cells subjected to oxygen glucose deprivation/reoxygenation (OGD/R) were used as CI/RI cell models. The expression of SNHG14, miR-98-5p and BCL2 like 13 (BCL2L13) was examined using quantitative real-time PCR (qPCR) or western blot. Apoptosis was monitored by flow cytometry assay. Apoptosis-related markers and endoplasmic reticulum (ER) stress-related markers were quantified by western blot. Inflammatory factors and oxidative stress were detected using matched commercial kits. The predicted relationship between miR-98-5p and SNHG14 or BCL2L13 was validated by dual-luciferase reporter assay, RIP assay and pull-down assay. RESULTS The high expression of SNHG14 was monitored in MCAO/R-treated mice and OGD/R-treated N2A cells. OGD/R-induced N2A cell apoptosis, ER stress, inflammation and oxidative stress were attenuated by SNHG14 knockdown. SNHG14 targeted miR-98-5p to positively regulate BCL2L13 expression. Inhibition of miR-98-5p recovered cell apoptosis, ER stress, inflammation and oxidative stress that were repressed by SNHG14 knockdown. Overexpression of BCL2L13 enhanced cell apoptosis, ER stress, inflammation and oxidative stress that were repressed by miR-98-5p enrichment. CONCLUSIONS SNHG14 knockdown alleviated OGD/induced N2A cell apoptosis, ER stress, inflammation and oxidative stress by depleting BCL2L13 via increasing miR-98-5p.
Collapse
Affiliation(s)
- Gaolian Zhang
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Jianhui Guo
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Jing Zeng
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Xiaoning Zhang
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Rui Chen
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Gang Wang
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China
| | - Weibin Liang
- Department of Craniocerebal, First Affiliated Hospital of Guangxi University of Chinese Medicine, No 327 Xianhu Avenue, 530023, Nanning, Guangxi, P. R. China.
| |
Collapse
|
42
|
Ding H, Wang K, He LQ, Yang XY, Huang HH, Liu WL, Xiao C, Du ZX, Yu L, Zhang W. A novel nano material for anti-cerebral ischemia: Preparation and Application of Borneol Angelica Polysaccharide Liposomes. J Liposome Res 2022:1-10. [DOI: 10.1080/08982104.2022.2106240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Huang Ding
- Hunan University of Chinese Medicine, Changsha, China
| | - Ke Wang
- Hunan University of Chinese Medicine, Changsha, China
| | - Lin-Qin He
- Hunan University of Chinese Medicine, Changsha, China
| | - Xiao-Yu Yang
- Hunan University of Chinese Medicine, Changsha, China
| | | | - Wen-Long Liu
- Hunan University of Chinese Medicine, Changsha, China
| | - Chen Xiao
- Hunan University of Chinese Medicine, Changsha, China
| | - Ze- Xuan Du
- Hunan University of Chinese Medicine, Changsha, China
| | - Lian- Yu
- Hunan University of Chinese Medicine, Changsha, China
| | - Wei Zhang
- Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
43
|
Zheng T, Lai X, Lu J, Chen Q, Wei D. Three Dimensional-Arterial Spin Labeling Evaluation of Improved Cerebral Perfusion After Limb Remote Ischemic Preconditioning in a Rat Model of Focal Ischemic Stroke. Front Neuroanat 2022; 16:893953. [PMID: 35847828 PMCID: PMC9280338 DOI: 10.3389/fnana.2022.893953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/06/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To investigate the application value of 3D arterial spin labeling (3D-ASL) for evaluating distal limb ischemic preconditioning to improve acute ischemic stroke (AIS) perfusion. Materials and Methods A total of 40 patients with AISs treated in our hospital from January 2020 to December 2020 were recruited, and 15 healthy individuals who were examined in our hospital during the same period were included as the control group; all of these participants were scored on the National Institutes of Health Stroke Scale (NIHSS) and examined by MRI. Sequences included conventional sequences, diffusion-weighted imaging (DWI), magnetic resonance angiography (MRA), and 3D-ASL, and cerebral infarct volume and cerebral blood flow (CBF) in the area of the infarct lesion were measured. After 3 months of treatment, patients with AIS were scored on the modified Rankin Scale (mRS) and divided into good prognosis and poor prognosis groups. In total, 55 adult male Sprague–Dawley rats were divided randomly into three groups: 20 in the middle cerebral artery occlusion (MCAO) group, 20 in the MCAO + limb remote ischemic preconditioning (LRP) group, and 15 in the sham group. In total, 48 h after the procedures, conventional MRI, DWI, and 3D-ASL sequence data were collected, and 2,3,5-trphenyltetrazolium chloride monohydrate (TTC) staining and behavioral scoring were performed. CBF was recorded in the infarct lesion area and the corresponding contralateral area, and the affected/contralateral relative values (rCBF) were calculated to compare the differences in rCBF between different groups. The pathological changes in brain tissues were observed by HE staining, and the expression of vascular endothelial growth factor (VEGF) and platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31) in brain tissues was detected by immunofluorescence and real-time quantitative polymerase chain reaction (RT-qPCR). The protein expression of VEGF was detected by western blotting. Results Hypertension and internal carotid atherosclerosis are high-risk factors for ischemic stroke, and CBF values in the infarct area are significantly lower than those in the corresponding areas on the contralateral side. NIHSS and mRS scores and CBF values have higher specificity and sensitivity for the prognosis of patients with AIS. LRP significantly reduces the infarct area, improves behavioral deficits in rats with cerebral ischemia, reduces neurological injury and histological damage, protects vascular structures, and promotes neovascularization. In addition, 3D-ASL showed a significant increase in brain tissue perfusion in the ischemic area after LRP, and the expression of VEGF and CD31 showed a significant positive correlation with CBF values. Conclusion Three dimensional (3D) ASL can be used to evaluate LRP to improve stroke perfusion, and its protective effect may be closely related to LRP-induced vascular regeneration.
Collapse
Affiliation(s)
- Tianxiu Zheng
- Department of Radiology, Ningde Municipal Hospital Affiliated to Ningde Normal University, Ningde, China
| | - Xiaolan Lai
- Department of Hematology, Ningde Municipal Hospital Affiliated to Ningde Normal University, Ningde, China
| | - Jiaojiao Lu
- Department of Central Laboratory, Ningde Municipal Hospital Affiliated to Ningde Normal University, Ningde, China
| | - Qiuyan Chen
- Department of Radiology, Ningde Municipal Hospital Affiliated to Ningde Normal University, Ningde, China
| | - Dingtai Wei
- Department of Radiology, Ningde Municipal Hospital Affiliated to Ningde Normal University, Ningde, China
- *Correspondence: Dingtai Wei,
| |
Collapse
|
44
|
Changes in Brain Electrical Activity after Transient Middle Cerebral Artery Occlusion in Rats. Neurol Int 2022; 14:547-560. [PMID: 35893279 PMCID: PMC9326608 DOI: 10.3390/neurolint14030044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/17/2022] [Accepted: 06/17/2022] [Indexed: 02/04/2023] Open
Abstract
Objectives. Ischemic stroke is a leading cause of death and disability worldwide. To search for new therapeutic and pharmacotherapeutic strategies, numerous models of this disease have been proposed, the most popular being transient middle cerebral artery occlusion. Behavioral and sensorimotor testing, biochemical, and histological methods are traditionally used in conjunction with this model to assess the effectiveness of potential treatment options. Despite its wide overall popularity, electroencephalography/electrocorticography is quite rarely used in such studies. Materials and methods. In the present work, we explored the changes in brain electrical activity at days 3 and 7 after 30- and 45-min of transient middle cerebral artery occlusion in rats. Results. Cerebral ischemia altered the amplitude and spectral electrocorticogram characteristics, and led to a reorganization of inter- and intrahemispheric functional connections. Ischemia duration affected the severity as well as the nature of the observed changes. Conclusions. The dynamics of changes in brain electrical activity may indicate a spontaneous partial recovery of impaired cerebral functions at post-surgery day 7. Our results suggest that electrocorticography can be used successfully to assess the functional status of the brain following ischemic stroke in rats as well as to investigate the dynamics of functional recovery.
Collapse
|
45
|
Guo H, Guo X, Jiang S. Long non-coding RNA lincRNA-erythroid prosurvival (EPS) alleviates cerebral ischemia/reperfusion injury by maintaining high-temperature requirement protein A1 (Htra1) stability through recruiting heterogeneous nuclear ribonucleoprotein L (HNRNPL). Bioengineered 2022; 13:12248-12260. [PMID: 35549989 PMCID: PMC9275866 DOI: 10.1080/21655979.2022.2074738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This study aimed at investigating the role and mechanism of lincRNA-EPS (erythroid prosurvival) in cerebral ischemia/reperfusion (CIR) injury. The results showed that the overexpression of lincRNA-EPS was able to reduce the levels of interleukin-6, tumor necrosis factor-alpha and interleukin-1β stimulated in the OGD-treated Neuro-2a (N-2a) cells. The levels of reactive oxygen species and malondialdehyde were enhanced while the superoxide dismutase levels were reduced by oxygen and glucose deprivation (OGD) treatment, in which the lincRNA-EPS overexpression could reverse this effect in the cells. LincRNA-EPS interacted with high-temperature requirement protein A1 (Htra1) and heterogeneous nuclear ribonucleoprotein L (HNRNPL), and their depletion inhibited the Htra1 mRNA stability in N-2a cells. HNRNPL knockdown blocked lincRNA-EPS overexpression-induced Htra1 expression in the cells. The depletion of Htra1 could rescue lincRNA-EPS overexpression-mediated N-2a cell injury, inflammation, and oxidative stress induced by OGD. Functionally, lincRNA-EPS alleviates CIR injury of the middle cerebral artery occlusion/reperfusion mice in vivo. In conclusion, lincRNA-EPS attenuates CIR injury by maintaining Htra1 stability through recruiting HNRNPL.
Collapse
Affiliation(s)
- Haifeng Guo
- Department of encephalopathy, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, Shandong, P.R.China
| | - Xia Guo
- Department of Obstetrics, Dongying People's Hospital, Dongying, Shandong, P.R.China
| | - Shiting Jiang
- Department of Internal Medicine-Neurology, Dongping People's Hospital, Taian, Shandong, P.R.China
| |
Collapse
|
46
|
Ji H, Jin H, Li G, Jin L, Ren X, Lv Y, Wang Y. Artemisinin protects against cerebral ischemia and reperfusion injury via inhibiting the NF-κB pathway. Open Med (Wars) 2022; 17:871-881. [PMID: 35950034 PMCID: PMC9096231 DOI: 10.1515/med-2022-0435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Abstract
This study investigated whether artemisinin (ART) exerts a neuroprotective effect against cerebral ischemia/reperfusion (I/R) injury. Hypoxia-glucose deprivation and reoxygenation (OGD/R) of SH-SY5Y cells were used as the I/R injury model in vitro. Cell viability was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and lactate dehydrogenase (LDH) release was measured. Cell apoptosis and apoptosis-associated protein expression were determined via flow cytometry and western blotting, respectively. The levels of glutathione peroxidase, superoxide dismutase, catalase, and malondialdehyde were determined. The secretion of tumor necrosis factor-α and interleukin-1β was measured using ELISA. The activation of the nuclear factor kappa B (NF-κB) pathway was also determined. The indicated ART concentrations (0, 25, 50, 75, and 100 μM) had no significant effect on SH-SY5Y cell viability and LDH activity. ART promoted cell viability, reduced cell apoptosis, repressed cellular inflammation, and inhibited cellular oxidative stress and NF-κB signaling pathway in OGD/R-induced SH-SY5Y cells. In addition, all the protective effects of ART on OGD/R-induced SH-SY5Y cell injury were significantly reversed by an NF-κB agonist. In conclusion, ART protects neurons from OGD/R-induced damage in vitro by inhibiting the NF-κB signaling pathway. These results suggest that ART may be a potential agent for the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Hui Ji
- Department of Basic Medicine, Qiqihar Medical University , Qiqihar , Heilongjiang 161006 , China
| | - Haifeng Jin
- Department of Basic Medicine, Qiqihar Medical University , Qiqihar , Heilongjiang 161006 , China
| | - Guangwei Li
- Department of Basic Medicine, Qiqihar Medical University , Qiqihar , Heilongjiang 161006 , China
| | - Li Jin
- Department of Basic Medicine, Qiqihar Medical University , Qiqihar , Heilongjiang 161006 , China
| | - Xiaoxu Ren
- Department of Basic Medicine, Qiqihar Medical University , Qiqihar , Heilongjiang 161006 , China
| | - Ying Lv
- Department of Basic Medicine, Qiqihar Medical University , Qiqihar , Heilongjiang 161006 , China
| | - Yuchun Wang
- College of Pharmacy, Qiqihar Medical University , Qiqihar , Heilongjiang 161006 , China
| |
Collapse
|
47
|
Roflumilast, a cyclic nucleotide phosphodiesterase 4 inhibitor, protects against cerebrovascular endothelial injury following cerebral ischemia/reperfusion by activating the Notch1/Hes1 pathway. Eur J Pharmacol 2022; 926:175027. [DOI: 10.1016/j.ejphar.2022.175027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/22/2022]
|
48
|
Artemisinin Alleviates Cerebral Ischemia/Reperfusion Injury via Regulation of the Forkhead Transcription Factor O1 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7824436. [PMID: 35422868 PMCID: PMC9005279 DOI: 10.1155/2022/7824436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/09/2022] [Accepted: 03/19/2022] [Indexed: 11/17/2022]
Abstract
The effect and mechanism of artemisinin therapy on cerebral ischemia-reperfusion injury (CIRI) was analyzed in this work. 100 healthy male C57BL/6 mice were selected and randomly divided into the sham group (no treatment), CIRI model group (IR), IR + artemisinin posttreatment group (IR + Arte), EX527 + IR group (EX527 + IR), and EX527 + IR + artemisinin posttreatment group (EX527 + IR + Arte), with 20 mice in each group. The cerebral infarct volumes of mice in different groups were measured by the 2,3,5-triphenyltetrazolium chloride (TTC) staining method. The neurological function scores and oxidative stress levels of mice in different groups were measured and compared. In addition, the expressions of silent information regulator 1 (SIRT1), forkhead transcription factor O1 (FOXO1), and p53 protein in brain tissue were detected. The results showed that the contents of reactive oxygen species (ROS) and malondialdehyde (MDA) in the EX527 + IR group and EX527 + IR + Arte group were significantly higher than those in the IR + Arte group (P < 0.05). The expressions of SIRT1 protein in the brain tissue of the IR group and EX527 + IR group were much lower than that of the sham group (P < 0.01); compared with the IR + Arte group, the expression of the X527 + IR group in the brain tissue was greatly reduced (P < 0.05). The expression levels of FOXO1 protein and p53 protein in the brain tissue of mice in the IR group and EX527 + IR group were higher than those in the sham group (P < 0.01). It was concluded that artemisinin treatment can reduce oxidative stress damage and alleviate CIRI through the SIRT1/FOXO1 signaling pathway, thereby achieving neuroprotective effects.
Collapse
|
49
|
Jiao Y, Wang J, Jia Y, Xue M. Remote ischemic preconditioning protects against cerebral ischemia injury in rats by upregulating miR-204-5p and activating the PINK1/Parkin signaling pathway. Metab Brain Dis 2022; 37:945-959. [PMID: 35067796 DOI: 10.1007/s11011-022-00910-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/12/2022] [Indexed: 11/25/2022]
Abstract
Remote ischemic preconditioning (RiPC) is the process where preconditioning ischemia protects the organs against the subsequent index ischemia. RiPC is a protective method for brain damage. This study is to explore the effect and mechanism of RiPC in cerebral ischemia injury in rats through regulation of miR-204-5p/BRD4 expression. Middle cerebral artery occlusion (MCAO) rat model and glucose deprivation (OGD) neuron model were established. The effect of RiPC on neurological deficits, cerebral infarct size, autophagy marker, inflammatory cytokines and apoptosis was evaluated. miR-204-5p expression was analyzed using RT-qPCR, and then downregulated using miR-204-5p antagomir to estimate its effect on MCAO rats. The downstream mechanism of miR-204-5p was explored. RiPC promoted autophagy, reduced cerebral infarct volume and neurological deficit score, and alleviated apoptosis and cerebral ischemia injury in rats, with no significant effects on healthy rat brains. RiPC up-regulated miR-204-5p expression in MCAO rats. miR-204-5p knockdown partially reversed the effect of RiPC. RiPC promoted autophagy in OGD cells, and attenuated inflammation and apoptosis. miR-204-5p targeted BRD4, which partially reversed the effect of miR-204-5p on OGD cells. RiPC activated the PINK1/Parkin pathway via the miR-204-5p/BRD4 axis. In conclusion, RiPC activated the PINK1/Parkin pathway and prevented cerebral ischemia injury by up-regulating miR-204-5p and inhibiting BRD4.
Collapse
Affiliation(s)
- Yiming Jiao
- The Departments of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - Jinlan Wang
- The Departments of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - Yanjie Jia
- The Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengzhou Xue
- The Departments of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China.
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China.
| |
Collapse
|
50
|
Akki R, Siracusa R, Cordaro M, Remigante A, Morabito R, Errami M, Marino A. Adaptation to oxidative stress at cellular and tissue level. Arch Physiol Biochem 2022; 128:521-531. [PMID: 31835914 DOI: 10.1080/13813455.2019.1702059] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several in vitro and in vivo investigations have already proved that cells and tissues, when pre-exposed to low oxidative stress by different stimuli such as chemical, physical agents and environmental factors, display more resistance against subsequent stronger ischaemic injuries, resulting in an adaptive response known as ischaemic preconditioning (IPC). The aim of this review is to report the most recent knowledge about the complex adaptive mechanisms, including signalling transduction pathways, antioxidant systems, apoptotic and inflammation pathways, underlying cell protection against oxidative damage. In addition, an update about in vivo adaptation strategies in response to ischaemic/reperfusion episodes and brain trauma is also given.
Collapse
Affiliation(s)
- Rachid Akki
- Department of Biology, Faculty of Science, University of Abdelmalek Essaadi, Tetouan, Morocco
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Remigante
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Rossana Morabito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Mohammed Errami
- Department of Biology, Faculty of Science, University of Abdelmalek Essaadi, Tetouan, Morocco
| | - Angela Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|