1
|
Bonová P, Končeková J, Némethová M, Morávek M, Baráth P, Danchenko M, Bona M. Remote ischemic conditioning-induced shift from a vulnerable to a tolerant penumbra: A proteomic perspective. Exp Neurol 2025; 391:115307. [PMID: 40381903 DOI: 10.1016/j.expneurol.2025.115307] [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: 02/28/2025] [Revised: 04/25/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
The concept of the ischaemic penumbra - stroke tissue with the potential to survive - has opened the door to a wide range of experimental strategies that could benefit the recovery of patients after a stroke. In this study, we used proteomic analysis to examine how remote ischaemic postconditioning (RIPC) mediates a shift from a vulnerable to a tolerant penumbra. We identified 450 differentially abundant proteins between the control group and the groups subjected to ischaemia via middle cerebral artery occlusion with or without RIPC during infarct expansion. The majority of proteins were downregulated following RIPC. Based on Gene Ontology enrichment analysis, we uncovered 24 gene sets significantly influenced during the reprogramming from a vulnerable to a tolerant penumbra. RIPC treatment positively impacted the synthesis of proteins enriched in the cytosol (GO:0005829) but inhibited the abundance of proteins belonging to the cytoskeleton (GO:0005874 microtubule) and the glutamatergic synapse (GO:0098978). The shift to a tolerant phenotype involved overexpression of aminopeptidases (GO:0004177) related to proteolysis (GO:0006508). RIPC also downregulated proteins involved in the tricarboxylic acid cycle (GO:0006099), adenosine triphosphate (ATP) binding (GO:0005524), and ATP hydrolysis (GO:0016887). We validated our proteomic findings by selecting two candidate genes (Map2 and Tubb3) for immunofluorescence. We identified the low-molecular-weight Map2 isoform as a potential marker of the shift from a vulnerable to a tolerant penumbra. In summary, our findings have revealed novel avenues for multimodal investigation of reprogramming the penumbra as part of recovery from stroke.
Collapse
Affiliation(s)
- Petra Bonová
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Košice, Slovak Republic.
| | - Jana Končeková
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Košice, Slovak Republic
| | - Miroslava Némethová
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Košice, Slovak Republic
| | - Marko Morávek
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Košice, Slovak Republic
| | - Peter Baráth
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 5807/9, 845 38 Bratislava, Slovak Republic
| | - Maksym Danchenko
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 5807/9, 845 38 Bratislava, Slovak Republic
| | - Martin Bona
- Department of Medical Physiology, Faculty of Medicine, University of Pavol Jozef Safarik, 040 01 Košice, Slovak Republic
| |
Collapse
|
2
|
Zhang H, Meléndez A. Conserved components of the macroautophagy machinery in Caenorhabditis elegans. Genetics 2025; 229:iyaf007. [PMID: 40180610 PMCID: PMC12005284 DOI: 10.1093/genetics/iyaf007] [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: 03/29/2024] [Accepted: 12/13/2024] [Indexed: 04/05/2025] Open
Abstract
Macroautophagy involves the sequestration of cytoplasmic contents in a double-membrane autophagosome and its subsequent delivery to lysosomes for degradation and recycling. In Caenorhabditis elegans, autophagy participates in diverse processes such as stress resistance, cell fate specification, tissue remodeling, aging, and adaptive immunity. Genetic screens in C. elegans have identified a set of metazoan-specific autophagy genes that form the basis for our molecular understanding of steps unique to the autophagy pathway in multicellular organisms. Suppressor screens have uncovered multiple mechanisms that modulate autophagy activity under physiological conditions. C. elegans also provides a model to investigate how autophagy activity is coordinately controlled at an organismal level. In this chapter, we will discuss the molecular machinery, regulation, and physiological functions of autophagy, and also methods utilized for monitoring autophagy during C. elegans development.
Collapse
Affiliation(s)
- Hong Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Alicia Meléndez
- Department of Biology, Queens College, City University of New York, Flushing, NY 11367, USA
- Molecular, Cellular and Developmental Biology and Biochemistry Ph.D. Programs, The Graduate Center of the City University of New York, New York, NY 10016, USA
| |
Collapse
|
3
|
Mukhamadeeva NR, Kachemaeva OV, Lakman IA, Buzaev IV, Galimova RM, Popova TA, Samorodov AV, Zagidullin NS. Neuroprotective effect of remote ischemic preconditioning in MRI-guided focused ultrasound treatment. RUSSIAN OPEN MEDICAL JOURNAL 2025; 14. [DOI: 10.15275/rusomj.2025.0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025] Open
Abstract
Introduction — Magnetic resonance imaging-guided focused ultrasound (MRgFUS) is used for ablation of various solid tumors and treatment of neurological diseases. During ablation, brain tissue is injured, resulting in the formation of necrosis and edema zones. Therefore, strategies to minimize brain damage are highly relevant. The goal of our research was to study the neuroprotective effect of remote ischemic preconditioning (RIPC) in the treatment of movement disorders using MRgFUS. Material and Methods — The study design was blinded randomized controlled 2-group trial. Patients were randomly assigned to the IPC group (systolic blood pressure +50 mm Hg, n=42) or sham IPC (sIPC) group (diastolic pressure, n=39) prior to MRgFUS in the mode of 3 cycles of 5 min of preconditioning on the forearm with 5 min of rest between cycles. Results — Following MRgFUS ablation of the ventralis intermediate (VIM) nucleus (gray matter), no differences in lesion size or tremor severity were detected after 2 h between the IPC and sIPC groups. When affecting the pallidothalamic tract (PTT, white matter), in the IPC group, in contrast to the sIPC, the main sizes of the necrotic lesion were significantly smaller after 2 h (frontal dimension, p=0.001; vertical dimension, p=0.037; necrosis lesion volume, p=0.007). Also, after 24 h in the IPC group, the dimensions of the lesion in the PTT were smaller (frontal dimension, p<0.001; vertical dimension, p=0.021; lesion volume, p=0.001). Conclusion — RIPC preceding MRgFUS has a neuroprotective effect on PTT (white matter).
Collapse
Affiliation(s)
| | - Olga V. Kachemaeva
- Bashkir State Medical University; V.S. Buzaev International Medical Center, Ufa, Russia
| | | | - Igor V. Buzaev
- Bashkir State Medical University; V.S. Buzaev International Medical Center, Ufa, Russia
| | - Rezida M. Galimova
- Bashkir State Medical University; V.S. Buzaev International Medical Center, Ufa, Russia
| | | | | | | |
Collapse
|
4
|
Li J, Liu S, Chen C, Deng J, Du Z, Yang S, Deng H, Zhang Z, Huang Y, Fu J, Zhang W, Poon WS, Hou H, Wang J. The Epileptiogenic Modified Therapy: Regulating the Dynamic of Microglia via ROS-Responsive Cascade Nano-Formulation. Adv Healthc Mater 2025; 14:e2403700. [PMID: 39713896 DOI: 10.1002/adhm.202403700] [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: 09/27/2024] [Revised: 12/13/2024] [Indexed: 12/24/2024]
Abstract
Pharmacological treatment of epilepsy presents several challenges, particularly the ineffectiveness of antiseizure medicines (ASMs) in modifying disease. In fact, the removal of reactive oxygen species (ROS) and preconditioning with tolerable dose of nitric oxide (NO) can activate neuroprotective mechanisms during latency and enhance tolerance to oxidative stress during seizures. To address this, a ROS-responsive cascade Nano-formulation (RRCN) is developed, which will transform ROS into NO. Remarkably, RRCN significantly reduces seizure severity, prolongs seizure latency, and extends inter-seizure intervals, though it does not increase the latency of generalized seizures. Microglia, the primary immune cells of the brain, play a crucial role in the initiation and progression of epilepsy. In the kainic acid (KA) epilepsy model, microglial processes elongate, branching increases, and interactions between microglia and neurons are strengthened in the CA1 and CA3 regions of the hippocampus compared to the Vehicle group. RRCN reverses these dynamic changes in microglia and their interactions with neurons, which are mediated by the NO/HIF/ErBb2 pathway. Thus, RRCN can inhibit seizures generalization by preconditioning the dynamic changes of microglia.
Collapse
Affiliation(s)
- Jiaxin Li
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chenghan Chen
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Jiahong Deng
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Zibo Du
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Simin Yang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Hongying Deng
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Zhixia Zhang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Yiyu Huang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Jingwen Fu
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Wangming Zhang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Wai Sang Poon
- Department of Surgery, Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jun Wang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
5
|
Wang J, Guan Z, Li W, Gong Y, Wang H, Zhou T, Liu J. The role of H3K27 acetylation in oxygen-glucose deprivation-induced spinal cord injury and potential for neuroprotective therapies. Brain Res Bull 2025; 220:111152. [PMID: 39643249 DOI: 10.1016/j.brainresbull.2024.111152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE Spinal cord injury (SCI) is a debilitating condition that often results in paralysis and lifelong medical challenges. Research has shown that epigenetic modifications, particularly histone acetylation, play a role in neuroprotection following hypoxic-ischemic events in SCI. The objective of this study was to explore the effects of histone H3K27 acetylation, along with its underlying mechanisms, on the tolerance to hypoxia and ischemia in SCI. METHODS This study employed an organotypic spinal cord slice culture model subjected to oxygen-glucose deprivation (OGD). We assessed cell apoptosis and changes in cellular type patterns under these conditions. Following hypoxia and ischemia, we analyzed the expression and distribution of H3K27ac across various nerve cell types. To identify key downstream genes, we integrated ChIP-seq and RNA-seq analyses, investigating molecular mechanisms driving the response to OGD in this model. RESULTS OGD stimulation increased cell apoptosis and induced time-dependent changes in the expression patterns of neurons, astrocytes, microglia, and oligodendrocytes in organotypic spinal cord slices, accompanied by a significant reduction in H3K27ac levels. Integrated ChIP-seq and RNA-seq analyses revealed that H3K27ac downregulation under hypoxic and ischemic conditions contributes to spinal cord damage by promoting neuroinflammation and disrupting gene regulation. Furthermore, we identified key downstream targets, including Apoc1, Spp1, Aff1, Brd4, KCNN3, and Rgma, which may represent promising therapeutic targets for SCI. CONCLUSION Our data underscore the pivotal role of H3K27ac in the organotypic spinal cord slice culture model following OGD exposure, offering promising avenues for neuroprotective therapies via epigenetic-immune regulation.
Collapse
Affiliation(s)
- Jing Wang
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Zheng Guan
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Weina Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Yu Gong
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Heying Wang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Ting Zhou
- Department of Laboratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Jingjie Liu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
6
|
Ntekoumes D, Song J, Liu H, Amelung C, Guan Y, Gerecht S. Acute Three-Dimensional Hypoxia Regulates Angiogenesis. Adv Healthc Mater 2025; 14:e2403860. [PMID: 39623803 PMCID: PMC11729260 DOI: 10.1002/adhm.202403860] [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: 10/06/2024] [Indexed: 01/15/2025]
Abstract
Hypoxia elicits a multitude of tissue responses depending on the severity and duration of the exposure. While chronic hypoxia is shown to impact development, regeneration, and cancer, the understanding of the threats of acute (i.e., short-term) hypoxia is limited mainly due to its transient nature. Here, a novel gelatin-dextran (Gel-Dex) hydrogel is established that decouples hydrogel formation and oxygen consumption and thus facilitates 3D sprouting from endothelial spheroids and, subsequently, induces hypoxia "on-demand." The Gel-Dex platform rapidly achieves acute moderate hypoxic conditions without compromising its mechanical properties. Acute exposure to hypoxia leads to increased endothelial cell migration and proliferation, promoting the total length and number of vascular sprouts. This work finds that the enhanced angiogenic response is mediated by reactive oxygen species, independently of hypoxia-inducible factors. Reactive oxygen species-dependent matrix metalloproteinases activity mediated angiogenic sprouting is observed following acute hypoxia. Overall, the Gel-Dex hydrogel offers a novel platform to study how "on-demand" acute moderate hypoxia impacts angiogenesis, with broad applicability to the development of novel sensing technologies.
Collapse
Affiliation(s)
- Dimitris Ntekoumes
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Haohao Liu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Connor Amelung
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Ya Guan
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
7
|
Song XY, Dong YH, Jia ZB, Chen LJ, Cui MY, Guan YJ, Yang BY, Wang SC, Chen SF, Li PK, Chen H, Zuo HC, Yang ZC, Xu WJ, Zhao YQ, Peng J. Protective effect of sub-hypothermic mechanical perfusion combined with membrane lung oxygenation on a yorkshire model of brain injury after traumatic blood loss. Chin J Traumatol 2024:S1008-1275(24)00175-5. [PMID: 39734147 DOI: 10.1016/j.cjtee.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 09/23/2024] [Accepted: 10/07/2024] [Indexed: 12/31/2024] Open
Abstract
PURPOSE To investigate the protective effect of sub-hypothermic mechanical perfusion combined with membrane lung oxygenation on ischemic hypoxic injury of yorkshire brain tissue caused by traumatic blood loss. METHODS This article performed a random controlled trial. Brain tissue of 7 yorkshire was selected and divided into the sub-low temperature anterograde machine perfusion group (n = 4) and the blank control group (n = 3) using the random number table method. A yorkshire model of brain tissue injury induced by traumatic blood loss was established. Firstly, the perfusion temperature and blood oxygen saturation were monitored in real-time during the perfusion process. The number of red blood cells, hemoglobin content, NA+, K+, and Ca2+ ions concentrations and pH of the perfusate were detected. Following perfusion, we specifically examined the parietal lobe to assess its water content. The prefrontal cortex and hippocampus were then dissected for histological evaluation, allowing us to investigate potential regional differences in tissue injury. The blank control group was sampled directly before perfusion. All statistical analyses and graphs were performed using GraphPad Prism 8.0 Student t-test. All tests were two-sided, and p value of less than 0.05 was considered to indicate statistical significance. RESULTS The contents of red blood cells and hemoglobin during perfusion were maintained at normal levels but more red blood cells were destroyed 3 h after the perfusion. The blood oxygen saturation of the perfusion group was maintained at 95% - 98%. NA+ and K+ concentrations were normal most of the time during perfusion but increased significantly at about 4 h. The Ca2+ concentration remained within the normal range at each period. Glucose levels were slightly higher than the baseline level. The pH of the perfusion solution was slightly lower at the beginning of perfusion, and then gradually increased to the normal level. The water content of brain tissue in the sub-low and docile perfusion group was 78.95% ± 0.39%, which was significantly higher than that in the control group (75.27% ± 0.55%, t = 10.49, p < 0.001), and the difference was statistically significant. Compared with the blank control group, the structure and morphology of pyramidal neurons in the Prefrontal cortex and CA1 region of the hippocampal gyrus were similar, and their integrity was better. The structural integrity of granulosa neurons was destroyed and cell edema increased in the perfusion group compared with the blank control group. Immunofluorescence staining for glail fibrillary acidic protein and Iba1, markers of glial cells, revealed well-preserved cell structures in the perfusion group. While there were indications of abnormal cellular activity, the analysis showed no significant difference in axon thickness or integrity compared to the 1-h blank control group. CONCLUSIONS Mild hypothermic machine perfusion can improve ischemia and hypoxia injury of yorkshire brain tissue caused by traumatic blood loss and delay the necrosis and apoptosis of yorkshire brain tissue by continuous oxygen supply, maintaining ion homeostasis and reducing tissue metabolism level.
Collapse
Affiliation(s)
- Xiang-Yu Song
- College of Clinical Medicine, Hebei University, Baoding, 071000, Hebei province, China
| | - Yang-Hui Dong
- Hebei North University, Graduate School, Zhangjiakou, 075051, Hebei province, China
| | - Zhi-Bo Jia
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Lei-Jia Chen
- Hebei North University, Graduate School, Zhangjiakou, 075051, Hebei province, China; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Meng-Yi Cui
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Yan-Jun Guan
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Medical School of PLA, Beijing, 100048, China
| | - Bo-Yao Yang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Medical School of PLA, Beijing, 100048, China
| | - Si-Ce Wang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Medical School of PLA, Beijing, 100048, China
| | - Sheng-Feng Chen
- Department of Orthopedics, The First Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Peng-Kai Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Medical School of PLA, Beijing, 100048, China
| | - Heng Chen
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Hao-Chen Zuo
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Zhan-Cheng Yang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Wen-Jing Xu
- Department of Orthopedics, The First Medical Center of Chinese PLA General Hospital, Beijing, 100048, China.
| | - Ya-Qun Zhao
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100048, China.
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China.
| |
Collapse
|
8
|
Wang L, Fu G, Han R, Fan P, Yang J, Gong K, Zhao Z, Zhang C, Sun K, Shao G. MALAT1 and NEAT1 are Neuroprotective During Hypoxic Preconditioning in the Mouse Hippocampus Possibly by Regulation of NR2B. High Alt Med Biol 2024; 25:285-294. [PMID: 38808452 DOI: 10.1089/ham.2023.0135] [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: 05/30/2024] Open
Abstract
Wang, Liping, Gang Fu, Ruijuan Han, Peijia Fan, Jing Yang, Kerui Gong, Zhijun Zhao, Chunyang Zhang, Kai Sun, and Guo Shao. MALAT1 and NEAT1 are neuroprotective during hypoxic preconditioning in the mouse hippocampus possibly by regulation of NR2B. High Alt Med Biol. 25:285-294, 2024. Background: The regulation of noncoding ribonucleic acid (ncRNA) has been shown to be involved in cellular and molecular responses to hypoxic preconditioning (HPC), a situation created by the induction of sublethal hypoxia in the brain. The ncRNAs metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and nuclear paraspeckle assembly transcript 1 (NEAT1) are abundantly expressed in the brain, where they regulate the expression of various genes in nerve cells. However, the exact roles of MALAT1 and NEAT1 in HPC are not fully understood. Methods: A mouse model of acute repeated hypoxia was used as a model of HPC, and MALAT1 and NEAT1 levels in the hippocampus were measured using real-time polymerase chain reaction (PCR). The mRNA and protein levels of N-methyl-d-aspartate receptor subunit 2 B (NR2B) in the mouse hippocampus were measured using real-time PCR and western blotting, respectively. HT22 cells knocked-down for MALAT1 and NEAT1 were used for in vitro testing. Expression of NR2B, which is involved in nerve cell injury under ischemic and hypoxic conditions, was also evaluated. The levels of spectrin and cleaved caspase-3 in MALAT1 and NEAT1 knockdown HT22 cells under oxygen glucose deprivation/reperfusion (OGD/R) were determined by western blotting. Results: HPC increased the expression of MALAT1 and NEAT1 and decreased the expression of NR2B mRNA in the mouse hippocampus (p < 0.05). Knockdown of MALAT1 and NEAT1 increased both NR2B mRNA and protein levels nearly twofold and caused damage under OGD/R conditions in HT22 cells (p < 0.05). Conclusion: MALAT1 and NEAT1 exert neuroprotective effects by influencing the expression of NR2B.
Collapse
Affiliation(s)
- Liping Wang
- Center for Translational Medicine, The Third People's Hospital of Longgang District, Shenzhen, PRC
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou, PRC
- Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Gang Fu
- Department of Cardiology, The Third People's Hospital of Longgang District, Shenzhen, PRC
| | - Ruijuan Han
- Department of Cardiology, The People's Hospital of Longgang District, Shenzhen, PRC
| | - Peijia Fan
- Zhongshan School of medicine, Sun Yat-sen University, Guangzhou, PRC
| | - Jing Yang
- Center for Translational Medicine, The Third People's Hospital of Longgang District, Shenzhen, PRC
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou, PRC
- Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
| | - Kerui Gong
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, USA
| | - Zhijun Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia, PRC
| | - Chunyang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia, PRC
| | - Kai Sun
- Center for Translational Medicine, The Third People's Hospital of Longgang District, Shenzhen, PRC
| | - Guo Shao
- Center for Translational Medicine, The Third People's Hospital of Longgang District, Shenzhen, PRC
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou, PRC
- Neuroscience Institute, Baotou Medical College, Inner Mongolia, PRC
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia, PRC
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PRC
- Joint Laboratory of South China Hospital Affiliated to Shenzhen University and Third People's Hospital of Longgang District, Shenzhen University, Shenzhen, PRC
| |
Collapse
|
9
|
Liu QY, Cui Y, Li W, Qiu J, Nguyen TN, Chen HS. Effect of remote ischemic preconditioning on cerebral circulation time in severe carotid artery stenosis: Results from the RIC-CCT trial. Cell Rep Med 2024; 5:101796. [PMID: 39471820 PMCID: PMC11604480 DOI: 10.1016/j.xcrm.2024.101796] [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: 07/10/2024] [Revised: 08/21/2024] [Accepted: 09/27/2024] [Indexed: 11/01/2024]
Abstract
In patients with severe internal carotid artery stenosis (sICAS), cerebral circulation time (CCT) is associated with cerebral hyperperfusion syndrome. This study aims to investigate the effect of remote ischemic preconditioning (RIC) on CCT in patients with sICAS. Patients are randomly assigned to the RIC group (RIC twice daily, for 2-4 days before carotid artery stenting [CAS] as an adjunct to standard medical therapy) and the control group. The results show that RIC produces a significant decrease in CCT of the stenosis side (sCCT) from baseline to pre-CAS, and the occurrence of contrast staining on brain computed tomography (CT) is lower in RIC versus control group after CAS. In addition, significant changes in some serum biomarkers suggest that anti-neuroinflammation, anti-oxidative stress, protecting endothelial injury, and improving cerebral autoregulation may be associated with the effect of RIC. These findings provide supporting evidence that RIC can modulate cerebral circulation in patients with sICAS. This study was registered at ClinicalTrials.gov (NCT05451030).
Collapse
Affiliation(s)
- Quan-Ying Liu
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Yu Cui
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Wei Li
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Jing Qiu
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Thanh N Nguyen
- Department of Neurology, Radiology, Boston Medical Center, Boston, MA, USA
| | - Hui-Sheng Chen
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China.
| |
Collapse
|
10
|
Zhu J, Xu N, Lin H, Deng L, Xie B, Jiang X, Liao R, Yang C. Remote ischemic preconditioning plays a neuroprotective role in cerebral ischemia-reperfusion mice by inhibiting mitophagy. Heliyon 2024; 10:e39076. [PMID: 39640619 PMCID: PMC11620096 DOI: 10.1016/j.heliyon.2024.e39076] [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: 03/14/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 12/07/2024] Open
Abstract
Remote ischemic preconditioning (RIPC) represents a clinically feasible method for safeguarding vital organs against ischemic injury. However, its specific role in cerebral ischemia-reperfusion (I/R) injury remains to be definitively elucidated. In this study, we investigated the neuroprotective effects of RIPC on mice at 7 days post-cerebral I/R and its involvement in mitophagy and mitochondrial dysfunction. Cerebral I/R led to impaired brain function, as well as structural and functional damage to mitochondria. Notably, RIPC treatment ameliorated the neurological dysfunction induced by cerebral I/R. Compared with the I/R group, the expression levels of NeuN, MBP, PDH, and Tom20 were significantly elevated in the RIPC + I/R group. Furthermore, mitochondria in the RIPC + I/R group exhibited more intact structure compared to those in the I/R group. In mice subjected to I/R injury, RIPC treatment markedly increased ATP content, ADP content, TAN level and glucose uptake while upregulating expression levels of Parkin, Pink1 and P62 proteins; it also reduced both the volume of ischemic foci and the number of mitochondrial autophagosomes along with decreasing LC3B II/I ratio. In conclusion, RIPC may exert a neuroprotective role by inhibiting excessive mitophagy during subacute stages following an ischemic stroke.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Na Xu
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Heng Lin
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoqian Jiang
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Runde Liao
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Chaoxian Yang
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Arslan R, Doganay S, Budak O, Bahtiyar N. Investigation of preconditioning and the protective effects of nicotinamide against cerebral ischemia-reperfusion injury in rats. Neurosci Lett 2024; 840:137949. [PMID: 39181500 DOI: 10.1016/j.neulet.2024.137949] [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: 02/27/2024] [Revised: 08/04/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
This study investigated the antioxidant and neuroprotective effects of nicotinamide combined with ischemic preconditioning against cerebral ischemia reperfusion (CIR) injury. Thirty-five Wistar albino male rats were randomly divided into five groups: sham, preconditioned ischemia/reperfusion (IP+IR), ischemia/reperfusion (IR), preconditioned ischemia/reperfusion + nicotinamide (IP+IR+N), and ischemia/reperfusion + nicotinamide (IR+N). CIR was achieved with bilateral common carotid artery occlusion. IP+IR and IP+IR+N groups 30 min before ischemia; Three cycles of 10 sec ischemia/30 sec reperfusion followed by 20 min IR were applied. The IP+IR+N and IR+N groups received 500 mg/kg nicotinamide intraperitoneally. After 24 h of reperfusion, a neurological evaluation was performed and vertıcal pole test. Biochemically, malondialdehyde (MDA), glutathione (GSH) levels and catalase (CAT) activity were measured in blood and brain tissue samples. Rates of red neurons, sateliosis and spongiosis were determined histopathologically in the prefrontal cortex areas. After CIR, MDA levels increased significantly in serum and brain tissue in the IR group compared to the sham group, while GSH and CAT activity decreased in the brain tissue (p < 0.05). MDA levels in the tissues were found significantly decreased in the IR+N group compared to the IR group (p < 0.05). Administration of nicotinamide together with IP significantly decreased MDA levels in brain tissue and increased GSH and CAT activity (p < 0.05). Compared to the IR group, the morphological and neurological damage in the prefrontal cortex areas decreased in the IP+IR, IP+IR+N, and IR+N groups (p < 0.05). In addition, red neuron, sateliosis and spongiosis rates increased significantly in the IR group compared to the Sham, IP+IR+N, IR+N groups (p < 0.001 for all). In neurological evaluation, while the neurological score increased and the time on the vertical pole decreased significantly in the IR group, preconditioning, and nicotinamide groups reversed (p < 0.05). The study's results show that nicotinamide administration with ischemic preconditioning alleviates cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ruhat Arslan
- Istinye University, Faculty of Medicine, Department of Physiology, TR - 34000 Istanbul, Turkey; Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Physiology, TR-34098 Istanbul, Turkey.
| | - Songul Doganay
- Sakarya University, Faculty of Medicine, Department of Physiology, TR-54000 Sakarya, Turkey.
| | - Ozcan Budak
- Sakarya University, Faculty of Medicine, Department of Histology and Embryology, TR-54000 Sakarya, Turkey.
| | - Nurten Bahtiyar
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Biophysics, TR-34098 Istanbul, Turkey.
| |
Collapse
|
12
|
Xinliang Z, Achkasov EE, Gavrikov LK, Yuchen L, Zhang C, Dudnik EN, Rumyantseva O, Beeraka NM, Glazachev OS. Assessing the importance and safety of hypoxia conditioning for patients with occupational pulmonary diseases: A recent clinical perspective. Biomed Pharmacother 2024; 178:117275. [PMID: 39126774 DOI: 10.1016/j.biopha.2024.117275] [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: 05/17/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Occupational pulmonary diseases (OPDs) pose a significant global health challenge, contributing to high mortality rates. This review delves into the pathophysiology of hypoxia and the safety of intermittent hypoxic conditioning (IHC) in OPD patients. By examining sources such as PubMed, Relemed, NLM, Scopus, and Google Scholar, the review evaluates the efficacy of IHC in clinical outcomes for OPD patients. It highlights the complexities of cardiovascular and respiratory regulation dysfunctions in OPDs, focusing on respiratory control abnormalities and the impact of intermittent hypoxic exposures. Key areas include the physiological effects of hypoxia, the role of hypoxia-inducible factor-1 alpha (HIF-1α) in occupational lung diseases, and the links between brain ischemia, stroke, and OPDs. The review also explores the interaction between intermittent hypoxic exposures, mitochondrial energetics, and lung physiology. The potential of IHE to improve clinical manifestations and underlying pathophysiology in OPD patients is thoroughly examined. This comprehensive analysis aims to benefit molecular pathologists, pulmonologists, clinicians, and physicians by enhancing understanding of IHE's clinical benefits, from research to patient care, and improving clinical outcomes for OPD patients.
Collapse
Affiliation(s)
- Zhang Xinliang
- Chair of Sports Medicine and Rehabilitation, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia; Co-Chair of Normal Physiology, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| | - Eugeny E Achkasov
- Chair of Sports Medicine and Rehabilitation, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| | - Leonid K Gavrikov
- Volgograd State Medical University, 1, Pavshikh Bortsov Sq., Volgograd 400131, Russia.
| | - Li Yuchen
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| | - Chen Zhang
- Chair of Epidemiology and Modern Technologies of Vaccination, Institute of Professional Education, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia
| | - Elena N Dudnik
- Co-Chair of Normal Physiology, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| | - Olga Rumyantseva
- Izmerov Research Institute of Occupational Health, 31 Budeynniy Avenye, Moscow 105275, Russia.
| | - Narasimha M Beeraka
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN 46202, USA; Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia; Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu, Anantapuramu, Andhra Pradesh 515721, India.
| | - Oleg S Glazachev
- Co-Chair of Normal Physiology, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| |
Collapse
|
13
|
Li H, Zhang X, Xu H, Liu H, Zhang Y, Zhang L, Zhou Y, Zhang Y, Liu J, Jing M, Zhang P, Yang P. Alternation of gene expression in brain-derived exosomes after cerebral ischemic preconditioning in mice. Heliyon 2024; 10:e35936. [PMID: 39224379 PMCID: PMC11367060 DOI: 10.1016/j.heliyon.2024.e35936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Aims Cerebral ischemic preconditioning is a neuroprotective therapy against cerebral ischemia and ischemia-reperfusion injury. This study aims to demonstrate the alternation of gene expression in exosomes from brain tissue of mice after ischemic preconditioning and their potential functions. Methods Ten mice were divided into the sham and the cerebral ischemic preconditioning groups. Their brain tissues were harvested, from which the exosomes were extracted. The characteristics and protective effects of exosomes were evaluated. Whole transcriptome sequencing was used to demonstrate the gene expression discrepancy between the exosomes from the two groups of mice brains. Volcano graphs and heatmaps were used to picture the difference in expression quantity of mRNA, lncRNA, and circRNA. Gene ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to demonstrate the functions of differentially expressed RNAs. Results Exosomes were successfully extracted, and those from the cerebral ischemic preconditioning group had better protective effects on cells that received oxygen-glucose deprivation and restoration injury. A total of 306 mRNAs and 374 lncRNAs were significantly upregulated, and 320 mRNAs and 405 lncRNAs were significantly downregulated in the preconditioning group. No circRNAs were differentially expressed between the two groups. GO and KEGG pathway analysis indicated that the functions of differentially expressed RNAs were related to both neural protective and injurious effects. Conclusion The brain-derived exosomes may participate in the neuroprotective effect of cerebral ischemic preconditioning. Thorough research is necessary to investigate exosome functions derived from the ischemic preconditioned brain.
Collapse
Affiliation(s)
- He Li
- Emergency Department, Naval Medical Center of PLA, Naval Medical University, Shanghai, China
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaoxi Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hongye Xu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hanchen Liu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yongxin Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lei Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yu Zhou
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yongwei Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianmin Liu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mei Jing
- Emergency Department, Naval Medical Center of PLA, Naval Medical University, Shanghai, China
| | - Ping Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Neurology, Naval Medical Center of PLA, Naval Medical University, Shanghai, China
| | - Pengfei Yang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
14
|
Wang Y, Kuca K, You L, Nepovimova E, Heger Z, Valko M, Adam V, Wu Q, Jomova K. The role of cellular senescence in neurodegenerative diseases. Arch Toxicol 2024; 98:2393-2408. [PMID: 38744709 PMCID: PMC11272704 DOI: 10.1007/s00204-024-03768-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024]
Abstract
Increasing evidence has revealed that cellular senescence drives NDs, including Alzheimer's disease (AD) and Parkinson's disease. Different senescent cell populations secrete senescence-associated secretory phenotypes (SASP), including matrix metalloproteinase-3, interleukin (IL)-1α, IL-6, and IL-8, which can harm adjacent microglia. Moreover, these cells possess high expression levels of senescence hallmarks (p16 and p21) and elevated senescence-associated β-galactosidase activity in in vitro and in vivo ND models. These senescence phenotypes contribute to the deposition of β-amyloid and tau-protein tangles. Selective clearance of senescent cells and SASP regulation by inhibiting p38/mitogen-activated protein kinase and nuclear factor kappa B signaling attenuate β-amyloid load and prevent tau-protein tangle deposition, thereby improving cognitive performance in AD mouse models. In addition, telomere shortening, a cellular senescence biomarker, is associated with increased ND risks. Telomere dysfunction causes cellular senescence, stimulating IL-6, tumor necrosis factor-α, and IL-1β secretions. The forced expression of telomerase activators prevents cellular senescence, yielding considerable neuroprotective effects. This review elucidates the mechanism of cellular senescence in ND pathogenesis, suggesting strategies to eliminate or restore senescent cells to a normal phenotype for treating such diseases.
Collapse
Affiliation(s)
- Yating Wang
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, 500 05, Hradec Kralove, Czech Republic
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| | - Li You
- College of Physical Education and Health, Chongqing College of International Business and Economics, Chongqing, 401520, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, 613 00, Brno, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, 613 00, Brno, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic.
| | - Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, 949 74, Nitra, Slovakia.
| |
Collapse
|
15
|
Mao Z, Zhang J, Guo L, Wang X, Zhu Z, Miao M. Therapeutic approaches targeting the gut microbiota in ischemic stroke: current advances and future directions. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:321-328. [PMID: 39364121 PMCID: PMC11444859 DOI: 10.12938/bmfh.2024-022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/02/2024] [Indexed: 10/05/2024]
Abstract
Ischemic stroke (IS) is the predominant form of stroke pathology, and its clinical management remains constrained by therapeutic time frame. The gut microbiota (GM), comprising a multitude of bacterial and archaeal cells, surpasses the human cell count by approximately tenfold and significantly contributes to the human organism's growth, development, and overall well-being. The microbiota-gut-brain axis (MGBA) in recent years has established a strong association between gut microbes and the brain, demonstrating their intricate involvement in the progression of IS. The regulation of IS by the GM, encompassing changes in composition, abundance, and distribution, is multifaceted, involving neurological, endocrine, immunological, and metabolic mechanisms. This comprehensive understanding offers novel insights into the therapeutic approaches for IS. The objective of this paper is to examine the mechanisms of interaction between the GM and IS in recent years, assess the therapeutic effects of the GM on IS through various interventions, such as dietary modifications, probiotics, fecal microbiota transplantation, and antibiotics, and offer insights into the potential clinical application of the GM in stroke treatment.
Collapse
Affiliation(s)
- Zhiguo Mao
- Department of Pharmacology, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan Province, China
| | - Jinying Zhang
- Department of Pharmacology, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan Province, China
| | - Lin Guo
- Department of Pharmacology, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan Province, China
| | - Xiaoran Wang
- Department of Pharmacology, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan Province, China
- The First Clinical Medical College, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan 450046, China
| | - Zhengwang Zhu
- The First Clinical Medical College, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan 450046, China
| | - Mingsan Miao
- Department of Pharmacology, Henan University of Chinese Medicine, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, No. 156 Jinshui East Road, Zhengdong New District, Zhengzhou, Henan Province, China
| |
Collapse
|
16
|
Zhang P, Fu G, Xu W, Gong K, Zhao Z, Sun K, Zhang C, Han R, Shao G. Up-regulation of miR-126 via DNA methylation in hypoxia-preconditioned endothelial cells may contribute to hypoxic tolerance of neuronal cells. Mol Biol Rep 2024; 51:808. [PMID: 39002003 DOI: 10.1007/s11033-024-09774-1] [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: 03/27/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Endothelial cells (ECs) can confer neuroprotection by secreting molecules. This study aimed to investigate whether DNA methylation contributes to the neuroprotective gene expression induced by hypoxia preconditioning (HPC) in ECs and to clarify that the secretion of molecules from HPC ECs may be one of the molecular mechanisms of neuroprotection. METHODS Human microvascular endothelial cell-1 (HMEC-1) was cultured under normal conditions (C), hypoxia(H), and hypoxia preconditioning (HPC), followed by the isolation of culture medium (CM). SY5Y cell incubated with the isolated CM from HMEC-1 was exposed to oxygen-glucose deprivation (OGD). The DNA methyltransferases (DNMTs), global methylation level, miR-126 and its promotor DNA methylation level in HMEC-1 were measured. The cell viability and cell injury in SY5Y were detected. RESULTS HPC decreased DNMTs level and global methylation level as well as increased miR-126 expression in HMEC-1. CM from HPC treated HMEC-1 also relieved SY5Y cell damage, while CM from HMEC-1 which over-expression of miR-126 can reduce injury in SY5Y under OGD condition. CONCLUSIONS These findings indicate EC may secrete molecules, such as miR-126, to execute neuroprotection induced by HPC through regulating the expression of DNMTs.
Collapse
Affiliation(s)
- Pu Zhang
- Center for Translational Medicine, the Third People's Hospital of Longgang District, Shenzhen, PR China
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou, PR China
| | - Gang Fu
- Department of Cardiology, the Third People's Hospital of Longgang District, Shenzhen, PR China
| | - Wenqing Xu
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou, PR China
| | - Kerui Gong
- Department of Oral and Maxillofacial Surgery, Department of Neurosurgery, University of California San Francisco, San Francisco, USA
| | - Zhujun Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, PR China
| | - Kai Sun
- Center for Translational Medicine, the Third People's Hospital of Longgang District, Shenzhen, PR China
| | - Chunyang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, PR China.
| | - Ruijuan Han
- Department of Cardiology, the Third People's Hospital of Longgang District, Shenzhen, PR China.
| | - Guo Shao
- Center for Translational Medicine, the Third People's Hospital of Longgang District, Shenzhen, PR China.
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou, PR China.
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, PR China.
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PR China.
- Joint Laboratory of South China Hospital Affiliated to Shenzhen University and Third, People's Hospital of Longgang District, Shenzhen University, Shenzhen, PR China.
| |
Collapse
|
17
|
Liu C, Liu G, Zuo X, Qu D, Sun Y, Liu L, Zhao X, Li J, Cai L. MiR-18a affects hypoxia induced glucose metabolism transition in HT22 hippocampal neuronal cell line through the Hif1a gene. BMC Neurol 2024; 24:204. [PMID: 38879468 PMCID: PMC11179257 DOI: 10.1186/s12883-024-03717-w] [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: 03/22/2024] [Accepted: 06/07/2024] [Indexed: 06/19/2024] Open
Abstract
Hypoxia can cause a variety of diseases, including ischemic stroke and neurodegenerative diseases. Within a certain range of partial pressure of oxygen, cells can respond to changes in oxygen. Changes in oxygen concentration beyond a threshold will cause damage or even necrosis of tissues and organs, especially for the central nervous system. Therefore, it is very important to find appropriate measures to alleviate damage. MiRNAs can participate in the regulation of hypoxic responses in various types of cells. MiRNAs are involved in regulating hypoxic responses in many types of tissues by activating the hypoxia-inducible factor (HIF) to affect angiogenesis, glycolysis and other biological processes. By analyzing differentially expressed miRNAs in hypoxia and hypoxia-related studies, as well as the HT22 neuronal cell line under hypoxic stress, we found that the expression of miR-18a was changed in these models. MiR-18a could regulate glucose metabolism in HT22 cells under hypoxic stress by directly regulating the 3'UTR of the Hif1a gene. As a small molecule, miRNAs are easy to be designed into small nucleic acid drugs, so this study can provide a theoretical basis for the research and treatment of nervous system diseases caused by hypoxia.
Collapse
Affiliation(s)
- Chuncheng Liu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou, 014010, China
| | - Gehui Liu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou, 014010, China
| | - Xinyang Zuo
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Donghui Qu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou, 014010, China
| | - Yefeng Sun
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Linan Liu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou, 014010, China
| | - Xiujuan Zhao
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou, 014010, China
| | - Jun Li
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou, 014010, China
| | - Lu Cai
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China.
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Baotou, 014010, China.
| |
Collapse
|
18
|
Ye T, Zhang N, Zhang A, Sun X, Pang B, Wu X. Electroacupuncture pretreatment alleviates rats cerebral ischemia-reperfusion injury by inhibiting ferroptosis. Heliyon 2024; 10:e30418. [PMID: 38807610 PMCID: PMC11130460 DOI: 10.1016/j.heliyon.2024.e30418] [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: 11/06/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Objective To explore the preventive effect of electroacupuncture pretreatment on stroke in rats by inhibiting ferroptosis and oxidative stress. Methods Rats were randomly assigned to the sham, middle cerebral artery occlusion/reperfusion (MCAO/R), MCAO/R + EP, MCAO/R + EP + erastin, and MCAO/R + EP + ferrostatin 1 groups. Daily electroacupuncture was performed 2 weeks before establishing the MCAO/R model utilizing the modified Zea Longa suture method. Rats were sacrificed 1 day after reperfusion, and brain tissues were collected. They were prepared for hematoxylin and eosin staining, prussian blue staining, transmission electron microscope. Measurement of total iron levels using a commercial kit, detection of malondialdehyde (MDA) and superoxide dismutase (SOD) levels by ELISA, and examination of 15-lox2, GPX4, SLC7A11, ACSL4, and TFR1 by western blotting. Results Compared with sham rats, cerebral infarction size was dramatically larger in MCAO/R rats. Moreover, the MCAO/R group displayed damaged mitochondria with a disarranged structure of cristae; free iron, total iron levels, and oxidative stress were significantly higher. Cerebral pathological lesions, oxidative stress, total iron levels, and protein levels of ACSL4, TFR1, and 15-lox2 were significantly reduced in the MCAO/R + EP and MCAO/R + EP + ferrostatin 1 groups, while the protective effect of electroacupuncture pretreatment on cerebral ischemia-reperfusion injury was inhibited by treatment with the ferroptosis activator erastin. Conclusion Electroacupuncture pretreatment can protect rats from cerebral ischemia-reperfusion injury by reducing the area of cerebral infarction and inhibiting ferroptosis and oxidative stress.
Collapse
Affiliation(s)
- Tao Ye
- Department of Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Ning Zhang
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Anbang Zhang
- Department of Neurology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Xiuqi Sun
- Department of Neurology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Bo Pang
- Department of Neurology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Xuemei Wu
- Department of Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| |
Collapse
|
19
|
Zhang L, Zhou X, Zhao J, Wang X. Research hotspots and frontiers of preconditioning in cerebral ischemia: A bibliometric analysis. Heliyon 2024; 10:e24757. [PMID: 38317957 PMCID: PMC10839892 DOI: 10.1016/j.heliyon.2024.e24757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/13/2023] [Accepted: 01/12/2024] [Indexed: 02/07/2024] Open
Abstract
Background Preconditioning is a promising strategy against ischemic brain injury, and numerous studies in vitro and in vivo have demonstrated its neuroprotective effects. However, at present there is no bibliometric analysis of preconditioning in cerebral ischemia. Therefore, a comprehensive overview of the current status, hot spots, and emerging trends in this research field is necessary. Materials and methods Studies on preconditioning in cerebral ischemia from January 1999-December 2022 were retrieved from the Web of Science Core Collection (WOSCC) database. CiteSpace was used for data mining and visual analysis. Results A total of 1738 papers on preconditioning in cerebral ischemia were included in the study. The annual publications showed an upwards and then downwards trend but currently remain high in terms of annual publications. The US was the leading country, followed by China, the most active country in recent years. Capital Medical University published the largest number of articles. Perez-Pinzon, Miguel A contributed the most publications, while KITAGAWA K was the most cited author. The focus of the study covered three areas: (1) relevant diseases and experimental models, (2) types of preconditioning and stimuli, and (3) mechanisms of ischemic tolerance. Remote ischemic preconditioning, preconditioning of mesenchymal stem cells (MSCs), and inflammation are the frontiers of research in this field. Conclusion Our study provides a visual and scientific overview of research on preconditioning in cerebral ischemia, providing valuable information and new directions for researchers.
Collapse
Affiliation(s)
- Long Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Traditional Chinese Medicine, Zibo TCM-Integrated Hospital, Zibo ,255026, China
| | - Xue Zhou
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jing Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xingchen Wang
- Division of Neurology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250001, China
| |
Collapse
|
20
|
Wang Y, Xu R, Yan Y, He B, Miao C, Fang Y, Wan H, Zhou G. Exosomes-Mediated Signaling Pathway: A New Direction for Treatment of Organ Ischemia-Reperfusion Injury. Biomedicines 2024; 12:353. [PMID: 38397955 PMCID: PMC10886966 DOI: 10.3390/biomedicines12020353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Ischemia reperfusion (I/R) is a common pathological process which occurs mostly in organs like the heart, brain, kidney, and lung. The injury caused by I/R gradually becomes one of the main causes of fatal diseases, which is an urgent clinical problem to be solved. Although great progress has been made in therapeutic methods, including surgical, drug, gene therapy, and transplant therapy for I/R injury, the development of effective methods to cure the injury remains a worldwide challenge. In recent years, exosomes have attracted much attention for their important roles in immune response, antigen presentation, cell migration, cell differentiation, and tumor invasion. Meanwhile, exosomes have been shown to have great potential in the treatment of I/R injury in organs. The study of the exosome-mediated signaling pathway can not only help to reveal the mechanism behind exosomes promoting reperfusion injury recovery, but also provide a theoretical basis for the clinical application of exosomes. Here, we review the research progress in utilizing various exosomes from different cell types to promote the healing of I/R injury, focusing on the classical signaling pathways such as PI3K/Akt, NF-κB, Nrf2, PTEN, Wnt, MAPK, toll-like receptor, and AMPK. The results suggest that exosomes regulate these signaling pathways to reduce oxidative stress, regulate immune responses, decrease the expression of inflammatory cytokines, and promote tissue repair, making exosomes a competitive emerging vector for treating I/R damage in organs.
Collapse
Affiliation(s)
- Yanying Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (Y.W.); (B.H.); (C.M.)
| | - Ruojiao Xu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| | - Yujia Yan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| | - Binyu He
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (Y.W.); (B.H.); (C.M.)
| | - Chaoyi Miao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (Y.W.); (B.H.); (C.M.)
| | - Yifeng Fang
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| | - Guoying Zhou
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China; (R.X.); (Y.Y.); (Y.F.)
| |
Collapse
|
21
|
Jackson CW, Xu J, Escobar I, Saul I, Fagerli E, Dave KR, Perez-Pinzon MA. Resveratrol Preconditioning Downregulates PARP1 Protein to Alleviate PARP1-Mediated Cell Death Following Cerebral Ischemia. Transl Stroke Res 2024; 15:165-178. [PMID: 36633794 PMCID: PMC10336177 DOI: 10.1007/s12975-022-01119-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023]
Abstract
Stroke remains a leading cause of mortality; however, available therapeutics are limited. The study of ischemic tolerance, in paradigms such as resveratrol preconditioning (RPC), provides promise for the development of novel prophylactic therapies. The heavily oxidative environment following stroke promotes poly-ADP-ribose polymerase 1 (PARP1)-overactivation and parthanatos, both of which are major contributors to neuronal injury. In this study, we tested the hypothesis that RPC instills ischemic tolerance through decreasing PARP1 overexpression and parthanatos following in vitro and in vivo cerebral ischemia. To test this hypothesis, we utilized rat primary neuronal cultures (PNCs) and middle cerebral artery occlusion (MCAO) in the rat as in vitro and in vivo models, respectively. RPC was administered 2 days preceding ischemic insults. RPC protected PNCs against oxygen and glucose deprivation (OGD)-induced neuronal loss, as well as increases in total PARP1 protein, implying protection against PARP1-overactivation. Twelve hours following OGD, we observed reductions in NAD+/NADH as well as an increase in AIF nuclear translocation, but RPC ameliorated NAD+/NADH loss and blocked AIF nuclear translocation. MCAO in the rat induced AIF nuclear translocation in the ischemic penumbra after 24 h, which was ameliorated with RPC. We tested the hypothesis that RPC's neuroprotection was instilled through long-term downregulation of nuclear PARP1 protein. RPC downregulated nuclear PARP1 protein for at least 6 days in PNCs, likely contributing to RPC's ischemic tolerance. This study describes a novel mechanism by which RPC instills prophylaxis against ischemia-induced PARP1 overexpression and parthanatos, through a long-term reduction of nuclear PARP1 protein.
Collapse
Grants
- R01 NS045676 NINDS NIH HHS
- 3R01NS034773, R01NS45676, R01NS054147 NIH HHS
- R01 NS054147 NINDS NIH HHS
- RF1 NS034773 NINDS NIH HHS
- R01 NS097658 NINDS NIH HHS
- R01 NS034773 NINDS NIH HHS
- 3R01NS034773, R01NS45676, R01NS054147 NIH HHS
- 3R01NS034773, R01NS45676, R01NS054147 NIH HHS
- 3R01NS034773, R01NS45676, R01NS054147 NIH HHS
- 3R01NS034773, R01NS45676, R01NS054147 NIH HHS
- 3R01NS034773, R01NS45676, R01NS054147 NIH HHS
Collapse
Affiliation(s)
- Charles W Jackson
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33136, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Jing Xu
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33136, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Iris Escobar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33136, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Isabel Saul
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33136, USA
| | - Eric Fagerli
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33136, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33136, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33136, USA.
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
22
|
Wang R, Yan Y, Tie Y, Zhang Q, Pan Y, Li S, Fan J, Li C, Li X, Wang Y, Sun X, Zhang T, Zhao X. Hypoxic acclimatization training improves the resistance to motion sickness. Front Neurosci 2023; 17:1216998. [PMID: 38125401 PMCID: PMC10731277 DOI: 10.3389/fnins.2023.1216998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Objective Vestibular provocation is one of the main causes of flight illusions, and its occurrence is closely related to the susceptibility of motion sickness (MS). However, existing training programs have limited effect in improving the resistance to motion sickness. In this study, we investigated the effects of hypoxia acclimatization training (HAT) on the resistance to motion sickness. Methods Healthy military college students were identified as subjects according to the criteria. MS model was induced by a rotary chair. Experimental groups included control, HAT, 3D roller training (3DRT), and combined training. Results The Graybiel scores were decreased in the HAT group and the 3DRT group and further decreased in the combined training group in MS induced by the rotary chair. Participants had a significant increase in blood pressure after the rotary chair test and a significant increase in the heart rate during the rotary chair test, but these changes disappeared in all three training groups. Additionally, LFn was increased, HFn was decreased, and LF/HF was increased accordingly during the rotary chair test in the control group, but the changes of these three parameters were completely opposite in the three training groups during the rotary chair test. Compared with the control group, the decreasing changes in pupillary contraction velocity (PCV) and pupillary minimum diameter (PMD) of the three training groups were smaller. In particular, the binocular PCV changes were further attenuated in the combined training group. Conclusion Our research provides a possible candidate solution for training military pilots in the resistance to motion sickness.
Collapse
Affiliation(s)
- Rui Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yiquan Yan
- Department of Aerospace Physiology, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yateng Tie
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Qi Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yikai Pan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Shuhan Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Jieyi Fan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Chengfei Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Xi Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yongchun Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Tongmei Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Xingcheng Zhao
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
23
|
Brock J, Basu N, Schlachetzki JCM, Schett G, McInnes IB, Cavanagh J. Immune mechanisms of depression in rheumatoid arthritis. Nat Rev Rheumatol 2023; 19:790-804. [PMID: 37923863 DOI: 10.1038/s41584-023-01037-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 11/06/2023]
Abstract
Depression is a common and disabling comorbidity in rheumatoid arthritis that not only decreases the likelihood of remission and treatment adherence but also increases the risk of disability and mortality in patients with rheumatoid arthritis. Compelling data that link immune mechanisms to major depressive disorder indicate possible common mechanisms that drive the pathology of the two conditions. Preclinical evidence suggests that pro-inflammatory cytokines, which are prevalent in rheumatoid arthritis, have various effects on monoaminergic neurotransmission, neurotrophic factors and measures of synaptic plasticity. Neuroimaging studies provide insight into the consequences of inflammation on the brain (for example, on neural connectivity), and clinical trial data highlight the beneficial effects of immune modulation on comorbid depression. Major depressive disorder occurs more frequently in patients with rheumatoid arthritis than in the general population, and major depressive disorder also increases the risk of a future diagnosis of rheumatoid arthritis, further highlighting the link between rheumatoid arthritis and major depressive disorder. This Review focuses on interactions between peripheral and central immunobiological mechanisms in the context of both rheumatoid arthritis and major depressive disorder. Understanding these mechanisms will provide a basis for future therapeutic development, not least in depression.
Collapse
Affiliation(s)
- James Brock
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Neil Basu
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Universitätsklinikum Erlangen, Erlangen, Germany
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Jonathan Cavanagh
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
24
|
Li Y, Fan Q, Li F, Pang R, Chen C, Li P, Wang X, Xuan W, Yu W. The multifaceted roles of activating transcription factor 3 (ATF3) in inflammatory responses - Potential target to regulate neuroinflammation in acute brain injury. J Cereb Blood Flow Metab 2023; 43:8-17. [PMID: 37165649 PMCID: PMC10638996 DOI: 10.1177/0271678x231171999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/22/2023] [Accepted: 04/03/2023] [Indexed: 05/12/2023]
Abstract
Activating transcription factor 3 (ATF3) is one of the most important transcription factors that respond to and exert dual effects on inflammatory responses. Recently, the involvement of ATF3 in the neuroinflammatory response to acute brain injury (ABI) has been highlighted. It functions by regulating neuroimmune activation and the production of neuroinflammatory mediators. Notably, recent clinical evidence suggests that ATF3 may serve as a potential ideal biomarker of the long-term prognosis of ABI patients. This mini-review describes the essential inflammation modulatory roles of ATF3 in different disease contexts and summarizes the regulatory mechanisms of ATF3 in the ABI-induced neuroinflammation.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyue Fan
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengshi Li
- Department of Neurosurgery, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Pang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Wang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Xuan
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Zhao BH, Ruze A, Zhao L, Li QL, Tang J, Xiefukaiti N, Gai MT, Deng AX, Shan XF, Gao XM. The role and mechanisms of microvascular damage in the ischemic myocardium. Cell Mol Life Sci 2023; 80:341. [PMID: 37898977 PMCID: PMC11073328 DOI: 10.1007/s00018-023-04998-z] [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: 05/22/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023]
Abstract
Following myocardial ischemic injury, the most effective clinical intervention is timely restoration of blood perfusion to ischemic but viable myocardium to reduce irreversible myocardial necrosis, limit infarct size, and prevent cardiac insufficiency. However, reperfusion itself may exacerbate cell death and myocardial injury, a process commonly referred to as ischemia/reperfusion (I/R) injury, which primarily involves cardiomyocytes and cardiac microvascular endothelial cells (CMECs) and is characterized by myocardial stunning, microvascular damage (MVD), reperfusion arrhythmia, and lethal reperfusion injury. MVD caused by I/R has been a neglected problem compared to myocardial injury. Clinically, the incidence of microvascular angina and/or no-reflow due to ineffective coronary perfusion accounts for 5-50% in patients after acute revascularization. MVD limiting drug diffusion into injured myocardium, is strongly associated with the development of heart failure. CMECs account for > 60% of the cardiac cellular components, and their role in myocardial I/R injury cannot be ignored. There are many studies on microvascular obstruction, but few studies on microvascular leakage, which may be mainly due to the lack of corresponding detection methods. In this review, we summarize the clinical manifestations, related mechanisms of MVD during myocardial I/R, laboratory and clinical examination means, as well as the research progress on potential therapies for MVD in recent years. Better understanding the characteristics and risk factors of MVD in patients after hemodynamic reconstruction is of great significance for managing MVD, preventing heart failure and improving patient prognosis.
Collapse
Affiliation(s)
- Bang-Hao Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Jing Tang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Nilupaer Xiefukaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Min-Tao Gai
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - An-Xia Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xue-Feng Shan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China.
| |
Collapse
|
26
|
Zhang Q, Zhao W, Li S, Ding Y, Wang Y, Ji X. Intermittent Hypoxia Conditioning: A Potential Multi-Organ Protective Therapeutic Strategy. Int J Med Sci 2023; 20:1551-1561. [PMID: 37859700 PMCID: PMC10583178 DOI: 10.7150/ijms.86622] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/08/2023] [Indexed: 10/21/2023] Open
Abstract
Severe hypoxia can induce a range of systemic disorders; however, surprising resilience can be obtained through sublethal adaptation to hypoxia, a process termed as hypoxic conditioning. A particular form of this strategy, known as intermittent hypoxia conditioning hormesis, alternates exposure to hypoxic and normoxic conditions, facilitating adaptation to reduced oxygen availability. This technique, originally employed in sports and high-altitude medicine, has shown promise in multiple pathologies when applied with calibrated mild to moderate hypoxia and appropriate hypoxic cycles. Recent studies have extensively investigated the protective role of intermittent hypoxia conditioning and its underlying mechanisms using animal models, demonstrating its potential in organ protection. This involves a range of processes such as reduction of oxidative stress, inflammation, and apoptosis, along with enhancement of hypoxic gene expression, among others. Given that intermittent hypoxia conditioning fosters beneficial physiological responses across multiple organs and systems, this review presents a comprehensive analysis of existing studies on intermittent hypoxia and its potential advantages in various organs. It aims to draw attention to the possibility of clinically applying intermittent hypoxia conditioning as a multi-organ protective strategy. This review comprehensively discusses the protective effects of intermittent hypoxia across multiple systems, outlines potential procedures for implementing intermittent hypoxia, and provides a brief overview of the potential protective mechanisms of intermittent hypoxia.
Collapse
Affiliation(s)
- Qihan Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenbo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Emergency Department, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Kong E, Li Y, Geng X, Wang J, He Y, Feng X. Ischemic preconditioning attenuates endoplasmic reticulum stress-dependent apoptosis of hepatocytes by regulating autophagy in hepatic ischemia-reperfusion injury. Int Immunopharmacol 2023; 122:110637. [PMID: 37473713 DOI: 10.1016/j.intimp.2023.110637] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/02/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
Hepatic ischemia-reperfusion injury (HIRI) usually occurs during subtotal hepatectomy and severely damages liver function during the perioperative period. Endoplasmic reticulum stress (ERS) dependent apoptosis has been suggested to play a crucial role in HIRI progression. The present study focused on the regulatory effect of autophagy activation induced by ischemic preconditioning (IPC) on ERS-dependent apoptosis of hepatocytes in HIRI. A HIRI mouse model and oxygen-glucose deprivation/reperfusion (OGD/R) AML-12 hepatocyte cell lines were constructed to evaluate the protective effect of IPC in vivo and in vitro. The protein levels of p-eIF2α, CHOP, and cleaved caspase-12 were used to evaluate the ERS-dependent apoptosis, whereas LC3-II and p62 were considered as the autophagy activation markers. The beneficial molecular chaperones GRP78, HSP60, and HSP70 were also tested to evaluate autophagy. HIRI significantly increased ERS-dependent apoptosis markers and the number of apoptotic cells and damaged liver function. The ERS inhibitor salubrinal significantly alleviated liver injury in HIRI and OGD/R hepatocytes. Furthermore, both remote IPC and direct IPC significantly alleviated liver injury and inflammatory cell infiltration. IPC also upregulated LC3-II, downregulated p62 expression, and increased the mRNA levels of GRP78, HSP60, and HSP70 in HIRI mice and OGD/R hepatocytes, indicating the activation of autophagy by IPC. The autophagy inhibitor 3-methyladenine significantly attenuated the protective effects of IPC on ERS-dependent apoptosis and liver function, whereas the autophagy activator rapamycin mimicked the protective effects of IPC on ERS-dependent apoptosis in vivo and in vitro, suggesting a regulatory role of autophagy in ERS-dependent apoptosis. These results demonstrated that IPC could induce moderate autophagy and upregulate a few molecular chaperones to strengthen endogenous defense mechanisms, which is beneficial for alleviating ERS-dependent apoptosis and protecting hepatocytes from HIRI.
Collapse
Affiliation(s)
- Erliang Kong
- Department of Anesthesiology, The 988th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Zhengzhou 450042, Henan, China; Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yongchang Li
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xuqiang Geng
- Department of Rheumatology and Immunology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jianxin Wang
- Department of Anesthesiology, The 988th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Zhengzhou 450042, Henan, China
| | - Yan He
- Department of Anesthesiology, Fuzhou Maternity and Child Health Care Hospital, Fuzhou 350000, Fujian, China.
| | - Xudong Feng
- Department of Anesthesiology, The 988th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Zhengzhou 450042, Henan, China.
| |
Collapse
|
28
|
Zhang S, Fu W, Jia X, Bade R, Liu X, Xie Y, Xie W, Jiang S, Shao G. Hypoxic Preconditioning Modulates BDNF and Its Signaling through DNA Methylation to Promote Learning and Memory in Mice. ACS Chem Neurosci 2023; 14:2320-2332. [PMID: 37289948 PMCID: PMC10289091 DOI: 10.1021/acschemneuro.3c00069] [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: 02/02/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Hypoxic preconditioning (HPC) as an endogenous mechanism can resist hypoxia/ischemia injury and exhibit protective effects on neurological function including learning and memory. Although underlying molecular mechanisms remain unclear, HPC probably regulates the expression of protective molecules by modulating DNA methylation. Brain-derived neurotrophic factor (BDNF) activates its signaling upon binding to the tropomyosin-related kinase B (TrkB) receptor, which is involved in neuronal growth, differentiation, and synaptic plasticity. Therefore, this study focused on the mechanism by which HPC regulates BDNF and BDNF/TrkB signaling through DNA methylation to influence learning and memory. Initially, the HPC model was established by hypoxia stimulations on ICR mice. We found that HPC downregulated the expression of DNA methyltransferase (DNMT) 3A and DNMT3B. Then, the upregulation of BDNF expression in HPC mice was generated from a decrease in DNA methylation of the BDNF gene promoter detected by pyrophosphate sequencing. Subsequently, upregulation of BDNF activated BDNF/TrkB signaling and ultimately improved learning and spatial memory in HPC mice. Moreover, after mice were intracerebroventricularly injected with the DNMT inhibitor, the restraint of DNA methylation accompanied by an increase of BDNF and BDNF/TrkB signaling was also discovered. Finally, we observed that the inhibitor of BDNF/TrkB signaling prevented HPC from ameliorating learning and memory in mice. However, the DNMT inhibitor promoted spatial cognition in mice. Thus, we suggest that HPC may upregulate BDNF by inhibiting DNMTs and decreasing DNA methylation of the BDNF gene and then activate BDNF/TrkB signaling to improve learning and memory in mice. This may provide theoretical guidance for the clinical treatment of cognitive dysfunction caused by ischemia/hypoxia disease.
Collapse
Affiliation(s)
- Shiji Zhang
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
| | - Weng Fu
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
| | - Xiaoe Jia
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
- School
of Basic Medicine and Forensic Sciences, Baotou Medical College, Baotou 014060, China
| | - Rengui Bade
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
- School
of Medical Technology and Anesthesia, Baotou Medical College of Neuroscience
Institute, Baotou Medical College, Baotou 014060, China
| | - Xiaolei Liu
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
| | - Yabin Xie
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
- School
of Medical Technology and Anesthesia, Baotou Medical College of Neuroscience
Institute, Baotou Medical College, Baotou 014060, China
| | - Wei Xie
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
- School
of Medical Technology and Anesthesia, Baotou Medical College of Neuroscience
Institute, Baotou Medical College, Baotou 014060, China
| | - Shuyuan Jiang
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
| | - Guo Shao
- Inner
Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou 014060, China
- Center
for Translational Medicine and Department of Laboratory Medicine, The Third People’s Hospital of Longgang District, Shenzhen 518112, China
| |
Collapse
|
29
|
Tang Y, Wei L, Wu Z, Xu P, Mo M. Parkinson's disease in a patient with GBA and LRRK2 covariants after acute hypoxic insult: a case report. BMC Neurol 2023; 23:226. [PMID: 37301871 PMCID: PMC10257258 DOI: 10.1186/s12883-023-03269-5] [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: 07/26/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND The glucocerebrosidase (GBA) and leucine-rich repeat kinase 2 (LRRK2) genes are associated with the risk of sporadic Parkinson's disease (PD). As an environmental factor, hypoxic insults may impair dopamine neurons in the substantia nigra and exacerbate PD symptoms. However, covariants of GBA and LRRK2 combined with hypoxic insults in clinical cases of Parkinsonism have not yet been reported. CASE PRESENTATION A 69-year-old male patient with PD and his relatives were clinically characterized and sequenced using the whole-exome technique. A novel covariant, c.1448 T > C (p. L483P, rs421016) on GBA and c.691 T > C (p. S231P, rs201332859) on LRRK2 were identified in this patient who first developed bradykinesia and rigidity in the neck at one month after an acute hypoxic insult during mountaineering. The patient presented with a mask-like face, festinating gait, asymmetric bradykinesia, and moderate rigidity. These symptoms were treated with levodopa and pramipexole, resulting in a 65% improvement in the Unified Parkinson's Disease Rating Scale (UPDRS) motor score. These parkinsonian symptoms persisted and developed with hallucinations, constipation, and rapid eye movement sleep behavior disorder. After 4 years, the patient exhibited a wearing-off phenomenon and died from pulmonary infection 8 years after disease onset. His parents, wife, and siblings were not diagnosed with PD, and his son carried p. L483P without Parkinsonism-like symptoms. CONCLUSIONS This is a case report of PD after hypoxic insult in a patient carrying a covariant of GBA and LRRK2. This study may help us understand the interaction between genetic and environmental factors in clinical PD.
Collapse
Affiliation(s)
- Yuting Tang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijian Wei
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhuohua Wu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingshu Mo
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
30
|
Shan Y, Wang L, Sun J, Chang S, Di W, Lv H. Exercise preconditioning attenuates cerebral ischemia-induced neuronal apoptosis, Th17/Treg imbalance, and inflammation in rats by inhibiting the JAK2/STAT3 pathway. Brain Behav 2023; 13:e3030. [PMID: 37143406 PMCID: PMC10275560 DOI: 10.1002/brb3.3030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Exercise preconditioning (EP) is essential for preventing ischemic stroke. Recent studies have shown that EP exerts neuroprotective effects in the cerebral ischemia-reperfusion injury model. Nonetheless, there have been few reports on the relationship between EP and the Th17/Treg balance. Moreover, it is unclear whether the JAK2/STAT3 pathway is responsible for the neuroprotective effect of EP. Therefore, we aimed to explore the impact of EP, other than the anti-inflammatory and antiapoptotic functions, on the Th17/Treg balance via the JAK2/STAT3 pathway in a middle cerebral artery occlusion (MCAO)-induced model. RESULTS Fifty rats were randomly allocated into five groups, including the sham group (n = 10), EP+sham group (n = 10), MCAO group (n = 10), EP+MCAO group (n = 10), and EP+MCAO+JAK2/STAT3 pathway agonist (coumermycin A1, CA1) group (n = 10). The results indicated that EP alleviated neurological deficits, reduced infarct volume, and ameliorated neuronal apoptosis induced by MCAO. Additionally, the MCAO-induced Th17/Treg imbalance could be rectified by EP. The decreased levels of IL-10 and Foxp3 and increased IL-17 and RORα in the MCAO group were reversed by EP treatment. Regarding inflammation, EP reduced the concentrations of IL-6 and IL-17 and elevated those of IL-10 and TGF-β. The neuroprotective effects of EP were accompanied by decreased phosphorylation of JAK2 and STAT3. Furthermore, CA1 pretreatment diminished all the beneficial effects of EP partially. CONCLUSION Our findings suggest that EP contributes to attenuating neuronal apoptosis, Th17/Treg imbalance, and inflammation induced by MCAO via inhibiting the JAK2/STAT3 pathway, indicating its therapeutic potential in ischemic stroke.
Collapse
Affiliation(s)
- Yuan Shan
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| | - Le Wang
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| | - Jingying Sun
- Central Research LaboratoryShaanxi Provincial People's HospitalXi'anChina
| | - Sha Chang
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| | - Wei Di
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| | - Hua Lv
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| |
Collapse
|
31
|
Li K, Lu M, Cui M, Wang X, Zheng Y. The Notch pathway regulates autophagy after hypoxic-ischemic injury and affects synaptic plasticity. Brain Struct Funct 2023; 228:985-996. [PMID: 37083721 DOI: 10.1007/s00429-023-02639-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/29/2023] [Indexed: 04/22/2023]
Abstract
Following neonatal hypoxic-ischemia (HI) injury, it is crucial factor to reconstruct neural circuit and maintain neural network homeostasis for neurological recovery. A dynamic balance between the synthesis and degradation of synaptic protein is required for maintaining synaptic plasticity. Protein degradation is facilitated by autophagy. This study aimed to investigate the regulation of synaptic structural plasticity by the Notch pathway, by assessing changes in Notch pathway activation and their effects on synaptic proteins and autophagy after HI injury. The study involved 48 male newborn Yorkshire piglets, each weighing 1.0-1.5 kg and 3 days old. They were randomly assigned to two groups: the HI group and the Notch pathway inhibitor + HI group (n = 24 per group). Each group was further divided into six subgroups according to HI duration (n = 4 per group): a control subgroup, and 0-6, 6-12, 12-24, 24-48, and 48-72 h subgroups. The expression of Notch pathway-related proteins, including Notch1, Hes1, and Notch intracellular domains, increased following HI injury. The expression of autophagy proteins increased at 0-6 h and 6-12 h post-HI. The expression of synaptic proteins, such as postsynaptic density protein 95 (PSD95) and synaptophysin, increased 6-12 h and 12-24 h after HI, respectively. Notably, the increased expression of these proteins was reversed by a Notch pathway inhibitor. Transmission electron microscopy revealed the presence of autophagosome structures in synapses. These findings shed light on the underlying mechanisms of neurological recovery after HI injury and may provide insights into potential therapeutic targets for promoting neural circuit reconstruction and maintaining neural network homeostasis.
Collapse
Affiliation(s)
- Kexin Li
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Meng Lu
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Mengxu Cui
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Xiaoming Wang
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China.
| | - Yang Zheng
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
32
|
Schiavi A, Salveridou E, Brinkmann V, Shaik A, Menzel R, Kalyanasundaram S, Nygård S, Nilsen H, Ventura N. Mitochondria hormesis delays aging and associated diseases in Caenorhabditis elegans impacting on key ferroptosis players. iScience 2023; 26:106448. [PMID: 37020951 PMCID: PMC10067770 DOI: 10.1016/j.isci.2023.106448] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 11/28/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Excessive iron accumulation or deficiency leads to a variety of pathologies in humans and developmental arrest in the nematode Caenorhabditis elegans. Instead, sub-lethal iron depletion extends C. elegans lifespan. Hypoxia preconditioning protects against severe hypoxia-induced neuromuscular damage across species but it has low feasible application. In this study, we assessed the potential beneficial effects of genetic and chemical interventions acting via mild iron instead of oxygen depletion. We show that limiting iron availability in C. elegans through frataxin silencing or the iron chelator bipyridine, similar to hypoxia preconditioning, protects against hypoxia-, age-, and proteotoxicity-induced neuromuscular deficits. Mechanistically, our data suggest that the beneficial effects elicited by frataxin silencing are in part mediated by counteracting ferroptosis, a form of non-apoptotic cell death mediated by iron-induced lipid peroxidation. This is achieved by impacting on different key ferroptosis players and likely via gpx-independent redox systems. We thus point to ferroptosis inhibition as a novel potential strategy to promote healthy aging.
Collapse
Affiliation(s)
- Alfonso Schiavi
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Eva Salveridou
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Vanessa Brinkmann
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Anjumara Shaik
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | | | - Sumana Kalyanasundaram
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ståle Nygård
- Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Natascia Ventura
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
33
|
Zhang Z, Liu C, Zhou X, Zhang X. The Critical Role of Sirt1 in Subarachnoid Hemorrhages: Mechanism and Therapeutic Considerations. Brain Sci 2023; 13:brainsci13040674. [PMID: 37190639 DOI: 10.3390/brainsci13040674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/28/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
The subarachnoid hemorrhage (SAH) is an important cause of death and long-term disability worldwide. As a nicotinamide adenine dinucleotide-dependent deacetylase, silent information regulator 1 (Sirt1) is a multipotent molecule involved in many pathophysiological processes. A growing number of studies have demonstrated that Sirt1 activation may exert positive effects on SAHs by regulating inflammation, oxidative stress, apoptosis, autophagy, and ferroptosis. Thus, Sirt1 agonists may serve as potential therapeutic drugs for SAHs. In this review, we summarized the current state of our knowledge on the relationship between Sirt1 and SAHs and provided an updated overview of the downstream molecules of Sirt1 in SAHs.
Collapse
Affiliation(s)
- Zhonghua Zhang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Cong Liu
- Department of Ophthalmology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
34
|
Li QY, Li XM, Hu HY, Ma YH, Ou YN, Wang AY, Tan L, Yu JT. Associations of Lung Function Decline with Risks of Cognitive Impairment and Dementia: A Meta-Analysis and Systematic Review. J Alzheimers Dis 2023; 92:853-873. [PMID: 36806509 DOI: 10.3233/jad-221136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
BACKGROUND There are controversies surrounding the effects of lung function decline on cognitive impairment and dementia. OBJECTIVE We conducted a meta-analysis and systematic review to explore the associations of lung function decline with the risks of cognitive impairment and dementia. METHODS The PubMed, EMBASE, and the Cochrane Library were searched to identify prospective studies published from database inception through January 10, 2023. We pooled relative risk (RR) and 95% confidence intervals (CI) using random-effects models. The Egger test, funnel plots, meta-regression, sensitivity, and subgroup analyses were conducted to detect publication bias and investigate the source of heterogeneity. RESULTS Thirty-three articles with a total of 8,816,992 participants were subjected to meta-analysis. Poorer pulmonary function was associated with an increased risk of dementia (FEV: RR = 1.25 [95% CI, 1.17-1.33]; FVC: RR = 1.40 [95% CI, 1.16-1.69]; PEF: RR = 1.84 [95% CI, 1.37-2.46]). The results of the subgroup analyses were similar to the primary results. Individuals with lung diseases had a higher combined risk of dementia and cognitive impairment (RR = 1.39 [95% CI, 1.20-1.61]). Lung disease conferred an elevated risk of cognitive impairment (RR = 1.37 [95% CI, 1.14-1.65]). The relationship between lung disease and an increased risk of dementia was only shown in total study participants (RR = 1.32 [95% CI, 1.11-1.57]), but not in the participants with Alzheimer's disease (RR = 1.39 [95% CI, 1.00-1.93]) or vascular dementia (RR = 2.11 [95% CI, 0.57-7.83]). CONCLUSION Lung function decline was significantly associated with higher risks of cognitive impairment and dementia. These findings might provide implications for the prevention of cognitive disorders and the promotion of brain health.
Collapse
Affiliation(s)
- Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Mei Li
- Department of Outpatient, Qingdao Municipal Hospital, Qingdao University, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - An-Yi Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Chen J, Xiao F, Chen L, Zhou Z, Wei Y, Zhong Y, Li L, Xie Y. Role of ferroptosis in hypoxic preconditioning to reduce propofol neurotoxicity. Front Pharmacol 2023; 14:1121280. [PMID: 36817119 PMCID: PMC9932196 DOI: 10.3389/fphar.2023.1121280] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Background: An increasing number of studies have reported that neurotoxicity of propofol may cause long-term learning and cognitive dysfunction. Hypoxic preconditioning has been shown to have neuroprotective effects, reducing the neurotoxicity of propofol. Ferroptosis is a new form of death that is different from apoptosis, necrosis, autophagy and pyroptosis. However, it is unclear whether hypoxic preconditioning reduces propofol neurotoxicity associated with ferroptosis. Thus, we aimed to evaluate the effect of propofol on primary hippocampal neurons in vitro to investigate the neuroprotective mechanism of hypoxic preconditioning and the role of ferroptosis in the reduction of propofol neurotoxicity by hypoxic preconditioning. Methods: Primary hippocampal neurons were cultured for 8 days in vitro and pretreated with or without propofol, hypoxic preconditioning, agonists or inhibitors of ferroptosis. Cell counting kit-8, Calcein AM, Reactive oxygen species (ROS), Superoxide dismutase (SOD), Ferrous iron (Fe2+), Malondialdehyde (MDA) and Mitochondrial membrane potential assay kit with JC-1 (JC-1) assays were used to measure cell viability, Reactive oxygen species level, Superoxide dismutase content, Fe2+ level, MDA content, and mitochondrial membrane potential. Cell apoptosis was evaluated using flow cytometry analyses, and ferroptosis-related proteins were determined by Western blot analysis. Results: Propofol had neurotoxic effects that led to decreased hippocampal neuronal viability, reduced mitochondrial membrane potential, decreased SOD content, increased ROS level, increased Fe2+ level, increased MDA content, increased neuronal apoptosis, altered expression of ferroptosis-related proteins and activation of ferroptosis. However, hypoxic preconditioning reversed these effects, inhibited ferroptosis caused by propofol and reduced the neurotoxicity of propofol. Conclusion: The neurotoxicity of propofol in developing rats may be related to ferroptosis. Propofol may induce neurotoxicity by activating ferroptosis, while hypoxic preconditioning may reduce the neurotoxicity of propofol by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Jing Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fei Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lifei Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhan Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yi Wei
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu Zhong
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Li
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China,*Correspondence: Yubo Xie, ; Li Li,
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China,Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China,*Correspondence: Yubo Xie, ; Li Li,
| |
Collapse
|
36
|
Guo M, Liu W, Luo H, Shao Q, Li Y, Gu Y, Guan Y, Ma W, Chen M, Yang H, Ji X, Liu J. Hypoxic stress accelerates the propagation of pathological alpha-synuclein and degeneration of dopaminergic neurons. CNS Neurosci Ther 2022; 29:544-558. [PMID: 36514210 PMCID: PMC9873519 DOI: 10.1111/cns.14055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/12/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022] Open
Abstract
AIMS The etiology of Parkinson's disease (PD) is complex and the mechanism is unclear. It has become a top priority to find common factors that induce and affect PD pathology. We explored the key role of hypoxia in promoting the pathological propagation of α-synuclein (α-syn) and the progression of PD. METHODS We performed PD modeling by conducting intracranial stereotaxic surgery in the unilateral striatum of mice. We then measured protein aggregation in vitro. The rotarod and pole tests were employed next to measure the damage of the phenotype. Pathological deposition and autophagy were also observed by immunofluorescence staining and protein levels measured by western blotting. RESULTS We demonstrated that short-term hypoxia activated phosphorylated (p)-α-syn in mice. We confirmed that p-α-syn was more readily formed aggregates than α-syn in vitro. Furthermore, we found that hypoxia promoted the activation and propagation of endogenous α-syn, contributing to the earlier degeneration of dopaminergic neurons in the substantia nigra and the deposition of p-α-syn in our animal model. Finally, autophagy inhibition contributed to the above pathologies. CONCLUSION Hypoxia was shown to accelerate the pathological progression and damage phenotype in PD model mice. The results provided a promising research target for determining common interventions for PD in the future.
Collapse
Affiliation(s)
- Mengyuan Guo
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Weijin Liu
- Department of Neurobiology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina,School of Rehabilitation MedicineCapital Medical UniversityBeijingChina
| | - Hanjiang Luo
- Neuroscience LaboratoryAffiliated Hospital of Guilin Medical UniversityGuangxiChina
| | - Qianqian Shao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yuning Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yakun Gu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Yuying Guan
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| | - Min Chen
- Neuroscience LaboratoryAffiliated Hospital of Guilin Medical UniversityGuangxiChina
| | - Hui Yang
- Department of Neurobiology, School of Basic Medical SciencesCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data‐based Precision MedicineCapital Medical UniversityBeijingChina
| |
Collapse
|
37
|
Li G, Liu J, Guo M, Gu Y, Guan Y, Shao Q, Ma W, Ji X. Chronic hypoxia leads to cognitive impairment by promoting HIF-2α-mediated ceramide catabolism and alpha-synuclein hyperphosphorylation. Cell Death Dis 2022; 8:473. [PMID: 36450714 PMCID: PMC9712431 DOI: 10.1038/s41420-022-01260-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022]
Abstract
Chronic hypoxia leads to irreversible cognitive impairment, primarily due to hippocampal neurodegeneration, for which the underlying mechanism remains poorly understood. We administered hypoxia (13%) to C57BL mice for 1-14 days in this study. Chronic hypoxia for 7 or 14 d, but not 1 or 3 d, resulted in alpha-synuclein hyperphosphorylation at serine129 (α-Syn p-S129) and protein aggregation, hippocampal neurodegeneration, and cognitive deficits, whereas the latter could be prevented by alpha-synuclein knockdown or an administered short peptide competing at α-Syn S129. These results suggest that α-Syn p-S129 mediates hippocampal degeneration and cognitive impairment following chronic hypoxia. Furthermore, we found that chronic hypoxia enhanced ceramide catabolism by inducing hypoxia-inducible factor (HIF)-2α and HIF-2α-dependent transcriptional activation of alkaline ceramidase 2 (Acer2). Thus, the enzymatic activity of protein phosphatase 2A (PP2A), a specific phosphatase for α-syn, is inhibited, leading to the sustained induction of α-Syn p-S129. Finally, we found that intermittent hypoxic preconditioning protected against subsequent chronic hypoxia-induced hippocampal neurodegeneration and cognitive impairment by preventing α-Syn p-S129. These results proved the critical role of α-syn pathology in chronic hypoxia-afforded cognitive impairment and revealed a novel mechanism underlying α-syn hyperphosphorylation during chronic hypoxia. The findings bear implications in developing novel therapeutic interventions for chronic hypoxia-related brain disorders.
Collapse
Affiliation(s)
- Gaifen Li
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China ,grid.413259.80000 0004 0632 3337Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Mengyuan Guo
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yakun Gu
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yuying Guan
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China ,grid.413259.80000 0004 0632 3337Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qianqian Shao
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Wei Ma
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China ,grid.413259.80000 0004 0632 3337Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
38
|
Yuan H, Liu J, Gu Y, Ji X, Nan G. Intermittent hypoxia conditioning as a potential prevention and treatment strategy for ischemic stroke: Current evidence and future directions. Front Neurosci 2022; 16:1067411. [PMID: 36507357 PMCID: PMC9732261 DOI: 10.3389/fnins.2022.1067411] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/11/2022] [Indexed: 11/26/2022] Open
Abstract
Ischemic stroke (IS) is the leading cause of disability and death worldwide. Owing to the aging population and unhealthy lifestyles, the incidence of cerebrovascular disease is high. Vascular risk factors include hypertension, diabetes, dyslipidemia, and obesity. Therefore, in addition to timely and effective reperfusion therapy for IS, it is crucial to actively control these risk factors to reduce the incidence and recurrence rates of IS. Evidence from human and animal studies suggests that moderate intermittent hypoxia (IH) exposure is a promising therapeutic strategy to ameliorate common vascular risk factors and comorbidities. Given the complex pathophysiological mechanisms underlying IS, effective treatment must focus on reducing injury in the acute phase and promoting repair in the recovery phase. Therefore, this review discusses the preclinical perspectives on IH conditioning as a potential treatment for neurovascular injury and highlights IH pre and postconditioning strategies for IS. Hypoxia conditioning reduces brain injury by increasing resistance to acute ischemic and hypoxic stress, exerting neuroprotective effects, and promoting post-injury repair and regeneration. However, whether IH produces beneficial effects depends not only on the hypoxic regimen but also on inter-subject differences. Therefore, we discuss the factors that may influence the effectiveness of IH treatment, including age, sex, comorbidities, and circadian rhythm, which can be used to help identify the optimal intervention population and treatment protocols for more accurate, individualized clinical translation. In conclusion, IH conditioning as a non-invasive, non-pharmacological, systemic, and multi-targeted intervention can not only reduce brain damage after stroke but can also be applied to the prevention and functional recovery of IS, providing brain protection at different stages of the disease. It represents a promising therapeutic strategy. For patients with IS and high-risk groups, IH conditioning is expected to develop as an adjunctive clinical treatment option to reduce the incidence, recurrence, disability, and mortality of IS and to reduce disease burden.
Collapse
Affiliation(s)
- Honghua Yuan
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yuhang Gu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China,*Correspondence: Xunming Ji,
| | - Guangxian Nan
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China,Guangxian Nan,
| |
Collapse
|
39
|
Schiavi A, Runci A, Maiorino T, Naso FD, Barenys M, Fritsche E, Strappazzon F, Ventura N. Cobalt chloride has beneficial effects across species through a hormetic mechanism. Front Cell Dev Biol 2022; 10:986835. [PMID: 36393859 PMCID: PMC9642780 DOI: 10.3389/fcell.2022.986835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/05/2022] [Indexed: 01/04/2025] Open
Abstract
Severe oxygen and iron deficiencies have evolutionarily conserved detrimental effects, leading to pathologies in mammals and developmental arrest as well as neuromuscular degeneration in the nematode Caenorhabditis elegans. Yet, similar to the beneficial effects of mild hypoxia, non-toxic levels of iron depletion, achieved with the iron chelator bipyridine or through frataxin silencing, extend C. elegans lifespan through hypoxia-like induction of mitophagy. While the positive health outcomes of hypoxia preconditioning are evident, its practical application is rather challenging. Here, we thus test the potential beneficial effects of non-toxic, preconditioning interventions acting on iron instead of oxygen availability. We find that limiting iron availability through the iron competing agent cobalt chloride has evolutionarily conserved dose-dependent beneficial effects: while high doses of cobalt chloride have toxic effects in mammalian cells, iPS-derived neurospheres, and in C. elegans, sub-lethal doses protect against hypoxia- or cobalt chloride-induced death in mammalian cells and extend lifespan and delay age-associated neuromuscular alterations in C. elegans. The beneficial effects of cobalt chloride are accompanied by the activation of protective mitochondrial stress response pathways.
Collapse
Affiliation(s)
- Alfonso Schiavi
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Alessandra Runci
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | | | | | - Marta Barenys
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Ellen Fritsche
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Flavie Strappazzon
- IRCCS Santa Lucia Foundation, Rome, Italy
- Institut NeuroMyogène, CNRS UMR5261—INSERM U1315, Université Claude Bernard Lyon1, Lyon, France
| | - Natascia Ventura
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| |
Collapse
|
40
|
Liu K, Cai Z, Zhang Q, He J, Cheng Y, Wei S, Yin M. Determination of significant parameters in remote ischemic postconditioning for ischemic stroke in experimental models: A systematic review and meta-analysis study. CNS Neurosci Ther 2022; 28:1492-1508. [PMID: 35896511 PMCID: PMC9437239 DOI: 10.1111/cns.13925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/04/2022] Open
Abstract
Objectives To systematically review studies using remote ischemia postconditioning (RIPostC) for ischemic stroke in experimental models and obtain factors that significantly influence treatment outcomes. Materials and Methods Peer‐reviewed studies were identified and selected based on the eligibility criteria, followed by extraction of data on potentially influential factors related to model preparation, postconditioning, and measure time based on outcome measures including infarct size, neurological scales, and cell tests with autophagy, apoptosis, normal‐neuron, and damaged‐neuron counting. Then, all data were preprocessed, grouped, and meta‐analyzed with the indicator of the standardized mean difference. Results Fifty‐seven studies with 224 experiments (91 for infarct size, 92 for neurological scales, and 41 for cell‐level tests) were included. There was little statistical difference between different model preparations, treated body parts, number of treatments, and sides. And treatment effect was generally a positive correlation with the duration of conditioning time to stroke onset with exceptions at some time points. Based on infarct size, the number of cycles per treatment, duration of occlusion, and release per cycle showed significant differences. Combined with the effect sizes by other measures, the occlusion/release duration of 8–10 min per cycle is better than 5 min, and three cycles per treatment were most frequently used with good effects. Effect also varied when measuring at different times, showing statistical differences in infarct size and most neurological scales. RIPostC is confirmed as an effective therapeutic intervention for ischemic stroke, while the RIPostC‐mediated autophagy level being activated or inhibited remained conflicting. Conclusions Conditioning time, number of cycles per treatment, duration of occlusion, and release per cycle were found to influence the treatment effects of RIPostC significantly. More studies on the relevant influential factors and autophagy mechanisms are warranted.
Collapse
Affiliation(s)
- Kezhou Liu
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| | - Zhengting Cai
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| | - Quanwei Zhang
- School of Management, Hangzhou Dianzi University, Hangzhou, China
| | - Jiatong He
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| | - Yinuo Cheng
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| | - Shaonong Wei
- HDU-ITMO Joint Institute, Hangzhou Dianzi University, Hangzhou, China
| | - Mengjie Yin
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| |
Collapse
|
41
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
42
|
Guan R, Yang C, Zhang J, Wang J, Chen R, Su P. Dehydroepiandrosterone alleviates hypoxia-induced learning and memory dysfunction by maintaining synaptic homeostasis. CNS Neurosci Ther 2022; 28:1339-1350. [PMID: 35703574 PMCID: PMC9344085 DOI: 10.1111/cns.13869] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/30/2022] Open
Abstract
AIMS Hypoxia causes plenty of pathologies in the central nervous system (CNS) including impairment of cognitive and memory function. Dehydroepiandrosterone (DHEA) has been proved to have therapeutic effects on CNS injuries by maintaining the homeostasis of synapses, yet its effect on hypoxia-induced CNS damage remains unknown. METHODS In vivo and in vitro models were established. Concentrations of glutamate and γ GABA were tested by ELISA. Levels of synapse-associated proteins were measured by western blotting. Density of dendritic protrusions of hippocampal neurons was assessed by Golgi staining. Immunofluorescence was adopted to observe the morphology of primary neurons. The novel object recognition test (NORT) and shuttle box test were used to evaluate cognition. RESULTS Dehydroepiandrosterone reversed abnormal elevation of glutamate levels, shortenings of neuronal processes, decreases in the density of dendritic protrusions, downregulation of synaptosome-associated protein (SNAP25), and impaired cognition caused by hypoxia. Hypoxia also resulted in notably downregulation of syntaxin 1A (Stx-1A). Overexpression of Stx-1A dramatically attenuated hypoxia-induced elevation of glutamate. Treatment with DHEA reversed the Stx-1A downregulation caused by hypoxic exposure. CONCLUSION Dehydroepiandrosterone may exert a protective effect on hypoxia-induced memory impairment by maintaining synaptic homeostasis. These findings offer a novel understanding of the therapeutic effect of DHEA on hypoxia-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Ruili Guan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Changhao Yang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Jianbin Zhang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Jianyu Wang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Peng Su
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
43
|
Jin X, Li P, Michalski D, Li S, Zhang Y, Jolkkonen J, Cui L, Didwischus N, Xuan W, Boltze J. Perioperative stroke: A perspective on challenges and opportunities for experimental treatment and diagnostic strategies. CNS Neurosci Ther 2022; 28:497-509. [PMID: 35224865 PMCID: PMC8928912 DOI: 10.1111/cns.13816] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/06/2023] Open
Abstract
Perioperative stroke is an ischemic or hemorrhagic cerebral event during or up to 30 days after surgery. It is a feared condition due to a relatively high incidence, difficulties in timely detection, and unfavorable outcome compared to spontaneously occurring stroke. Recent preclinical data suggest that specific pathophysiological mechanisms such as aggravated neuroinflammation contribute to the detrimental impact of perioperative stroke. Conventional treatment options are limited in the perioperative setting due to difficult diagnosis and medications affecting coagulation in may cases. On the contrary, the chance to anticipate cerebrovascular events at the time of surgery may pave the way for prevention strategies. This review provides an overview on perioperative stroke incidence, related problems, and underlying pathophysiological mechanisms. Based on this analysis, we assess experimental stroke treatments including neuroprotective approaches, cell therapies, and conditioning medicine strategies regarding their potential use in perioperative stroke. Interestingly, the specific aspects of perioperative stroke might enable a more effective application of experimental treatment strategies such as classical neuroprotection whereas others including cell therapies may be of limited use. We also discuss experimental diagnostic options for perioperative stroke augmenting classical clinical and imaging stroke diagnosis. While some experimental stroke treatments may have specific advantages in perioperative stroke, the paucity of established guidelines or multicenter clinical research initiatives currently limits their thorough investigation.
Collapse
Affiliation(s)
- Xia Jin
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | | | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yueman Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Jukka Jolkkonen
- Department of Neurology and A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lili Cui
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Nadine Didwischus
- School of Life Sciences, University of Warwick, Coventry, UK.,Department of Radiology, University of Pittsburgh, Pittsburgh, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
44
|
Zhu Y, Sun Y, Hu J, Pan Z. Insight Into the Mechanism of Exercise Preconditioning in Ischemic Stroke. Front Pharmacol 2022; 13:866360. [PMID: 35350755 PMCID: PMC8957886 DOI: 10.3389/fphar.2022.866360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/21/2022] [Indexed: 01/07/2023] Open
Abstract
Exercise preconditioning has attracted extensive attention to induce endogenous neuroprotection and has become the hotspot in neurotherapy. The training exercise is given multiple times before cerebral ischemia, effectively inducing ischemic tolerance and alleviating secondary brain damage post-stroke. Compared with other preconditioning methods, the main advantages of exercise include easy clinical operation and being readily accepted by patients. However, the specific mechanism behind exercise preconditioning to ameliorate brain injury is complex. It involves multi-pathway and multi-target regulation, including regulation of inflammatory response, oxidative stress, apoptosis inhibition, and neurogenesis promotion. The current review summarizes the recent studies on the mechanism of neuroprotection induced by exercise, providing the theoretical basis of applying exercise therapy to prevent and treat ischemic stroke. In addition, we highlight the various limitations and future challenges of translational medicine from fundamental study to clinical application.
Collapse
Affiliation(s)
- Yuanhan Zhu
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yulin Sun
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Jichao Hu
- Department of Orthopedics, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Zhuoer Pan
- Department of Orthopedics, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
45
|
Amantea D, La Russa D, Frisina M, Giordano F, Di Santo C, Panno ML, Pignataro G, Bagetta G. Ischemic Preconditioning Modulates the Peripheral Innate Immune System to Promote Anti-Inflammatory and Protective Responses in Mice Subjected to Focal Cerebral Ischemia. Front Immunol 2022; 13:825834. [PMID: 35359933 PMCID: PMC8962743 DOI: 10.3389/fimmu.2022.825834] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/21/2022] [Indexed: 11/26/2022] Open
Abstract
The development of tolerance triggered by a sublethal ischemic episode (preconditioning, PC) involves a complex crosstalk between neurons, astrocytes and microglia, although the role of the peripheral immune system in this context is largely unexplored. Here, we report that severe cerebral ischemia caused by transient middle cerebral artery occlusion (MCAo) in adult male mice elevates blood counts of inflammatory neutrophils and monocytes, and plasma levels of miRNA-329-5p. These inflammatory responses are prevented by ischemic PC induced by 15 min MCAo, 72h before the severe insult (1h MCAo). As compared with sham-operated animals, mice subjected to either ischemic PC, MCAo or a combination of both (PC+MCAo) display spleen contraction. However, protein levels of Ym1 (a marker of polarization of myeloid cells towards M2/N2 protective phenotypes) are elevated only in spleen from the experimental groups PC and PC+MCAo, but not MCAo. Conversely, Ym1 protein levels only increase in circulating leukocytes from mice subjected to 1h MCAo, but not in preconditioned animals, which is coincident with a dramatic elevation of Ym1 expression in the ipsilateral cortex. By immunofluorescence analysis, we observe that expression of Ym1 occurs in amoeboid-shaped myeloid cells, mainly representing inflammatory monocytes/macrophages and neutrophils. As a result of its immune-regulatory functions, ischemic PC prevents elevation of mRNA levels of the pro-inflammatory cytokine interleukin (IL)-1β in the ipsilateral cortex, while not affecting IL-10 mRNA increase induced by MCAo. Overall, the elevated anti-inflammatory/pro-inflammatory ratio observed in the brain of mice pre-exposed to PC is associated with reduced brain infarct volume and ischemic edema, and with amelioration of functional outcome. These findings reaffirm the crucial and dualistic role of the innate immune system in ischemic stroke pathobiology, extending these concepts to the context of ischemic tolerance and underscoring their relevance for the identification of novel therapeutic targets for effective stroke treatment.
Collapse
Affiliation(s)
- Diana Amantea
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Daniele La Russa
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Marialaura Frisina
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Chiara Di Santo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Giuseppe Pignataro
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, “Federico II” University, Naples, Italy
| | - Giacinto Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| |
Collapse
|
46
|
Introducing the brain erythropoietin circle to explain adaptive brain hardware upgrade and improved performance. Mol Psychiatry 2022; 27:2372-2379. [PMID: 35414656 PMCID: PMC9004453 DOI: 10.1038/s41380-022-01551-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
Abstract
Executive functions, learning, attention, and processing speed are imperative facets of cognitive performance, affected in neuropsychiatric disorders. In clinical studies on different patient groups, recombinant human (rh) erythropoietin (EPO) lastingly improved higher cognition and reduced brain matter loss. Correspondingly, rhEPO treatment of young rodents or EPO receptor (EPOR) overexpression in pyramidal neurons caused remarkable and enduring cognitive improvement, together with enhanced hippocampal long-term potentiation. The 'brain hardware upgrade', underlying these observations, includes an EPO induced ~20% increase in pyramidal neurons and oligodendrocytes in cornu ammonis hippocampi in the absence of elevated DNA synthesis. In parallel, EPO reduces microglia numbers and dampens their activity and metabolism as prerequisites for undisturbed EPO-driven differentiation of pre-existing local neuronal precursors. These processes depend on neuronal and microglial EPOR. This novel mechanism of powerful postnatal neurogenesis, outside the classical neurogenic niches, and on-demand delivery of new cells, paralleled by dendritic spine increase, let us hypothesize a physiological procognitive role of hypoxia-induced endogenous EPO in brain, which we imitate by rhEPO treatment. Here we delineate the brain EPO circle as working model explaining adaptive 'brain hardware upgrade' and improved performance. In this fundamental regulatory circle, neuronal networks, challenged by motor-cognitive tasks, drift into transient 'functional hypoxia', thereby triggering neuronal EPO/EPOR expression.
Collapse
|
47
|
Li G, Liu J, Guan Y, Ji X. The role of hypoxia in stem cell regulation of the central nervous system: From embryonic development to adult proliferation. CNS Neurosci Ther 2021; 27:1446-1457. [PMID: 34817133 PMCID: PMC8611781 DOI: 10.1111/cns.13754] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is involved in the regulation of various cell functions in the body, including the regulation of stem cells. The hypoxic microenvironment is indispensable from embryonic development to the regeneration and repair of adult cells. In addition to embryonic stem cells, which need to maintain their self-renewal properties and pluripotency in a hypoxic environment, adult stem cells, including neural stem cells (NSCs), also exist in a hypoxic microenvironment. The subventricular zone (SVZ) and hippocampal dentate gyrus (DG) are the main sites of adult neurogenesis in the brain. Hypoxia can promote the proliferation, migration, and maturation of NSCs in these regions. Also, because most neurons in the brain are non-regenerative, stem cell transplantation is considered as a promising strategy for treating central nervous system (CNS) diseases. Hypoxic treatment also increases the effectiveness of stem cell therapy. In this review, we firstly describe the role of hypoxia in different stem cells, such as embryonic stem cells, NSCs, and induced pluripotent stem cells, and discuss the role of hypoxia-treated stem cells in CNS diseases treatment. Furthermore, we highlight the role and mechanisms of hypoxia in regulating adult neurogenesis in the SVZ and DG and adult proliferation of other cells in the CNS.
Collapse
Affiliation(s)
- Gaifen Li
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jia Liu
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
| | - Yuying Guan
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|