1
|
Guan Y, Pan L, Niu D, Li X, Li S, Cheng G, Zeng Z, Yue R, Yao J, Zhang G, Sun C, Yang H. Mailuo Shutong pills inhibit neuroinflammation by regulating glucose metabolism disorders to protect mice from cerebral ischemia-reperfusion injury. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118621. [PMID: 39053718 DOI: 10.1016/j.jep.2024.118621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mailuo Shutong Pill (MLST), a traditional Chinese medicine (TCM), has been widely used for clearing heat and detoxifying, eliminating stasis and dredging meridians, dispelling dampness and diminishing swelling. Earlier study found that MLST could improve cerebral ischemic-reperfusion injury, however, the potential mechanism has not been well evaluated. AIM OF STUDY In this study, a well established and widely used mice model of middle cerebral artery occlusion/reperfusion (MCAO/R) was preformed to evaluate the protective function of MLST on cerebral ischemic-reperfusion injury and further discuss the potential pharmacological mechanisms. MATERIALS AND METHODS Chemical profiling of MLST was analyzed based on Ultra-high-performance liquid chromatography electrospray ionization orbitrap tandem mass spectrometry. ICR mice were challenged by MCAO/R surgery. The protective effect of MLST on MCAO/R injury was evaluated by neurological deficit score, cerebral infarct rate, brain water content, H&E and nissl staining. The blood-brain barrier (BBB) integrity was detected by Evans blue staining. The potential pharmacological mechanism of MLST in treating MCAO/R injury was further elucidated by the methods of proteomics, central carbon targeted metabolomics, as well as Western blot. Immunohistochemistry was used to detect the microglia infiltration, enzyme linked immunosorbent assay (ELISA) kit was explored to evaluate the content of IL-1β, TNF-α and IL-6 in brain tissue, and Western blot was used to detect proteins expression in brain tissue. RESULTS A total of 76 chemical compounds have been determined in MLST. MLST effectively protected mice from MCAO/R injury, which was confirmed by lower neurological deficit score, cerebral infarct rate, brain water content and nissl body loss, and improved brain pathology. Meanwhile, MLST upregulated the expression of ZO-1, Occludin and Claudin 5 by downregulating the ratio of TIMP1/MMP9 to suppress the entrance of Evans blue to brain tissue, indicating that MLST maintained the integrity of BBB. Further studies indicated that MLST inhibited the inflammatory level of brain tissue by inhibiting microglia infiltration and downregulating NLRP3 inflammasome signaling pathway. The results of proteomics, Western blot, and central carbon targeted metabolomics confirmed that MLST regulated Glycolysis/Gluconogenesis, Pyruvate metabolism and TCA cycle in brain tissue of mice with MCAO/R. CONCLUSION MLST inhibits neuroinflammation by regulating glucose metabolism disorders to interfere with immune metabolism reprogramming and inhibit the NLRP3 inflammasome signaling pathway, and finally improve cerebral ischemia-reperfusion injury. This study confirms that MLST is a potential drug for treating Cerebral ischemic stroke.
Collapse
Affiliation(s)
- Yongxia Guan
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Xin Li
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Shirong Li
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Guoliang Cheng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Rujing Yue
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Guimin Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Chenghong Sun
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China; College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, China.
| | - Hongjun Yang
- Changchun University of Chinese Medicine, Changchun, 130117, China; Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
2
|
Riessland M, Ximerakis M, Jarjour AA, Zhang B, Orr ME. Therapeutic targeting of senescent cells in the CNS. Nat Rev Drug Discov 2024; 23:817-837. [PMID: 39349637 DOI: 10.1038/s41573-024-01033-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 11/01/2024]
Abstract
Senescent cells accumulate throughout the body with advanced age, diseases and chronic conditions. They negatively impact health and function of multiple systems, including the central nervous system (CNS). Therapies that target senescent cells, broadly referred to as senotherapeutics, recently emerged as potentially important treatment strategies for the CNS. Promising therapeutic approaches involve clearing senescent cells by disarming their pro-survival pathways with 'senolytics'; or dampening their toxic senescence-associated secretory phenotype (SASP) using 'senomorphics'. Following the pioneering discovery of first-generation senolytics dasatinib and quercetin, dozens of additional therapies have been identified, and several promising targets are under investigation. Although potentially transformative, senotherapies are still in early stages and require thorough testing to ensure reliable target engagement, specificity, safety and efficacy. The limited brain penetrance and potential toxic side effects of CNS-acting senotherapeutics pose challenges for drug development and translation to the clinic. This Review assesses the potential impact of senotherapeutics for neurological conditions by summarizing preclinical evidence, innovative methods for target and biomarker identification, academic and industry drug development pipelines and progress in clinical trials.
Collapse
Affiliation(s)
- Markus Riessland
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| | | | | | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miranda E Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Salisbury VA Medical Center, Salisbury, NC, USA.
| |
Collapse
|
3
|
von Zglinicki T. Oxidative stress and cell senescence as drivers of ageing: Chicken and egg. Ageing Res Rev 2024; 102:102558. [PMID: 39454760 DOI: 10.1016/j.arr.2024.102558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Oxidative stress and cell senescence are both important drivers of ageing and age-associated disease and disability. In vitro, they are closely interconnected in a chicken-and-egg relationship: Not only is oxidative stress an important cause of cell senescence, but senescent cells are also sources of oxidative stress, obscuring cause-effect relationships during the ageing process. We hypothesize that cell senescence is a significant cause of tissue and systemic oxidative stress during ageing. This review aims to critically summarize the available evidence for this hypothesis. After summarizing the cellular feedback mechanisms that make oxidative stress an integral part of the senescent phenotype, it critically reviews the existing evidence for a role of senescent cells as causes of oxidative stress during mammalian ageing in vivo, focussing on results from intervention experiments. It is concluded that while the available data are in agreement with this hypothesis, they are still too scarce to support a robust conclusion.
Collapse
Affiliation(s)
- Thomas von Zglinicki
- Ageing Research Laboratories, Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Health, Newcastle University, UK.
| |
Collapse
|
4
|
Ni C, Huang B, Huang Y, Wen Z, Luo S. Keap1-independent GSK-3β/Nrf2 signaling mediates electroacupuncture inhibition of oxidative stress to induce cerebral ischemia-reperfusion tolerance. Brain Res Bull 2024; 217:111071. [PMID: 39241893 DOI: 10.1016/j.brainresbull.2024.111071] [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: 06/11/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE Cerebral ischemia-reperfusion (CIR) injury is a devastating consequence of stroke characterized by oxidative stress-induced neuronal damage. Electroacupuncture (EA) has emerged as a potential therapeutic intervention for ischemic stroke, but its underlying mechanisms remain incompletely understood. This study aimed to elucidate whether EA exerts anti-oxidative stress effects against CIR injury by modulating the GSK-3β/Nrf2 pathway. METHODS CIR mouse models were established using the suture-occluded method and underwent EA pretreatment. Cognitive and neurologic function, cerebral infarct volume, and neuronal damage were assessed in mice. Oxidative stress levels and the expression of components of the GSK-3β/Nrf2 pathway in the cerebral cortex were measured. The regulatory effect of GSK-3β on Nrf2 and its role in electroacupuncture to alleviate oxygen-glucose deprivation/reoxygenation (OGD/R)-induced neuronal injury were investigated by modulating GSK-3β expression in HT22 hippocampal neuronal cells and electroacupuncture serum intervention. Ultimately, Nrf2 knockout mice, GSK-3β knockout mice, and wild-type mice treated with TBHQ (an Nrf2 activator) were utilized for further validation. RESULTS EA pretreatment improved cognitive impairment and neuronal damage induced by CIR injury. Mechanistically, EA inhibited oxidative stress in the cerebral cortex, manifested by reduced levels of reactive oxygen species and malondialdehyde, along with increased superoxide dismutase activity. Furthermore, EA upregulated the expression of Nrf2 and its downstream antioxidant enzymes HO-1 and NQO1, while Keap1 expression remained unaffected. In vitro, GSK-3β overexpression inhibited the protective effects of EA serum on OGD/R-induced neuronal damage. In vivo, knockout of either Nrf2 or Gsk-3β genes abolished the neuroprotective effects of EA, and TBHQ exerted effects similar to EA, confirming the significant role of GSK-3β/Nrf2 in mediating EA antioxidative effects. CONCLUSION EA exerts antioxidative stress effects against CIR injury by activating the GSK-3β/Nrf2 signaling pathway, independent of Keap1 regulation.
Collapse
Affiliation(s)
- Chunjue Ni
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Baojun Huang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yufan Huang
- Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhengde Wen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Wenzhou Key Laboratory of Perioperative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Shan Luo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Wenzhou Key Laboratory of Perioperative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
5
|
You J, Li X, Xia J, Li H, Wang J. Hypoperfusion Intensity Ratio and Hemorrhagic Transformation in Patients with Successful Recanalization after Thrombectomy. AJNR Am J Neuroradiol 2024; 45:1475-1481. [PMID: 38719611 PMCID: PMC11448998 DOI: 10.3174/ajnr.a8329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/29/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND PURPOSE Hemorrhagic transformation remains a potentially devastating complication of acute ischemic stroke. We aimed to evaluate whether the hypoperfusion intensity ratio, a parameter derived from CT perfusion imaging, is associated with the development of hemorrhagic transformation in patients with anterior large-artery occlusion who had undergone thrombectomy. MATERIALS AND METHODS We retrospectively reviewed data from patients with consecutive acute ischemic strokes who had achieved successful recanalization (Thrombolysis in Cerebral Infarction score ≥2b) between January 2020 and December 2023. HIR was defined as the ratio of the volume of lesions with a time-to-maximum (Tmax) >6 seconds to those with a Tmax >10 second delay. The primary outcome, based on the European Cooperative Acute Stroke Study, was hemorrhagic transformation, diagnosed by follow-up imaging assessment in 24-hour windows, and radiologically classified as hemorrhagic infarction and parenchymal hematoma. The secondary outcome was a 3-month mRS score of ≥3. RESULTS Among 168 patients, 35 of 168 developed hemorrhagic transformation; 14 of 168 developed hemorrhagic infarction, and 21 of 168 developed parenchymal hematoma PH. After adjusting the latent covariates, increased hypoperfusion intensity ratio (per 0.1, adjusted OR [aOR] 1.68, 95% CI 1.26-2.25), ASPECTS (aOR 0.44, 95% CI 0.27-0.72), onset-to-puncture (aOR 1.01, 95% CI 1.00-1.02), and cardioembolism (aOR 5.6, 95% CI 1.59-19.7) were associated with hemorrhagic transformation in multivariable regression. The receiver operating characteristic curve indicated that hypoperfusion intensity ratio can predict hemorrhagic transformation accurately (area under the curve = 0.81; 95% CI, 0.738-0.882; P < .001) and predict parenchymal hematoma (area under the curve = 0.801; 95% CI, 0.727-0.875; P < .001). CONCLUSIONS Upon admission, hypoperfusion intensity ratio, an imaging parameter, predicted hemorrhagic transformation after reperfusion therapy in this patient population.
Collapse
Affiliation(s)
- Jiaxiang You
- From the Department of Emergency Medicine (J.Y., J.X., H.L., J.W.), Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China
| | - Xiaoxi Li
- Department of Emergency Medicine (X.L., J.W.), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jun Xia
- From the Department of Emergency Medicine (J.Y., J.X., H.L., J.W.), Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China
| | - Haopeng Li
- From the Department of Emergency Medicine (J.Y., J.X., H.L., J.W.), Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China
| | - Jun Wang
- From the Department of Emergency Medicine (J.Y., J.X., H.L., J.W.), Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China
- Department of Emergency Medicine (X.L., J.W.), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
6
|
Ou C, Lin Y, Wen J, Zhang H, Xu Y, Zhang N, Liu Q, Wu Y, Xu J, Wu J. Roflumilast Attenuates Microglial Senescence and Retinal Inflammatory Neurodegeneration Post Retinal Ischemia Reperfusion Injury Through Inhibiting NLRP3 Inflammasome. Invest Ophthalmol Vis Sci 2024; 65:38. [PMID: 39446353 PMCID: PMC11512574 DOI: 10.1167/iovs.65.12.38] [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: 05/12/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Purpose Retinal ischemia-reperfusion (RIR) injury is implicated in various retinal diseases, leading to retinal ganglion cells (RGCs) degeneration. Microglial senescence exacerbates inflammation, contributing to neurodegeneration. This study aimed to investigate the potential therapeutic role of Roflumilast (Roflu) in ameliorating microglial senescence and neuroinflammation following RIR injury. Methods C57BL/6J mice underwent RIR surgery, and Roflu treatment was administered intraperitoneally. BV2 microglial cells were subjected to oxygen-glucose deprivation and reoxygenation (OGD/R) to simulate ischemic conditions in vitro. SA-β-gal staining was used to detect cellular senescence. Quantitative PCR and ELISA were used to examine the levels of senescence-associated secretory phenotype (SASP) factors. Hematoxylin and eosin (H&E) staining was performed on retinal sections to assess retinal morphology and thickness. Surviving RGCs were labeled and quantified in retinal whole-mounts using immunofluorescence (IF). Furthermore, Western blot and IF staining were used to quantify the proteins associated with the cell cycle and NLRP3 inflammasomes. Results Roflu treatment reduced microglial senescence, ROS production, and secretion of pro-inflammatory cytokines in OGD/R-exposed BV2 cells. It also restored cell proliferation capacity and reversed OGD/R-induced cell cycle arrest. In vivo, Roflu alleviated retinal senescence, preserved retinal thickness, and protected against RGCs death in the RIR mouse model. Mechanistically, Roflu inhibited the NLRP3 inflammasome activation and suppressed DNA damage signaling pathway in microglia. Conclusions Roflu exerts neuroprotective effects by mitigating microglial senescence and inflammation via inhibition of the NLRP3 inflammasome in RIR injury. These findings suggest that Roflu may serve as a promising therapeutic strategy for retinal diseases associated with ischemic injury by targeting microglial senescence.
Collapse
Affiliation(s)
- Chunlian Ou
- Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of General Practice, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yiwei Lin
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Wen
- Department of Ophthalmology, Taizhou Central Hospital, Taizhou, Zhejiang, China
| | - Hongyang Zhang
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Xu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong, China
| | - Naiyuan Zhang
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiong Liu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yingzi Wu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Xu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Wu
- Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Sui C, Liu Y, Jiang J, Tang J, Yu L, Lv G. Ginsenoside Rg1 ameliorates cerebral ischemia-reperfusion injury by regulating Pink1/ Parkin-mediated mitochondrial autophagy and inhibiting microglia NLRP3 activation. Brain Res Bull 2024; 216:111043. [PMID: 39134096 DOI: 10.1016/j.brainresbull.2024.111043] [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: 02/21/2024] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024]
Abstract
OBJECTIVE This study aimed to further elucidate the mechanism of ginsenoside Rg1 in the treatment of cerebral ischemia-reperfusion. METHODS In this study, we observed the apoptosis of RM cells (microglia) after oxygen-glucose deprivation/reoxygenation (OGD/R) modeling before and after Rg1 administration, changes in mitochondrial membrane potential, changes in the content of Reactive oxygen species (ROS) and inflammatory vesicles NLR Family Pyrin Domain Containing 3 (NLRP3), and the expression levels of autophagy-related proteins, inflammatory factors, and apoptosis proteins. We further examined the pathomorphological changes in brain tissue, neuronal damage, changes in mitochondrial morphology and mitochondrial structure, and the autophagy-related proteins, inflammatory factors, and apoptosis proteins expression levels in CI/RI rats before and after administration of Rg1 in vivo experiments. RESULTS In vitro experiments showed that Rg1 induced mitochondrial autophagy, decreased mitochondrial membrane potential, and reduced ROS content thereby inhibiting NLRP3 activation, decreasing secretion of inflammatory factors and RM cell apoptosis by regulating the PTEN induced putative kinase 1(Pink1) /Parkin signaling pathway. In vivo experiments showed that Rg1 induced mitochondrial autophagy, inhibited NLRP3 activation, improved inflammatory response, and reduced apoptosis by regulating the Pink1/Parkin signaling pathway, and Rg1 significantly reduced the area of cerebral infarcts, improved the pathological state of brain tissue, and attenuated the neuronal damage, thus improving cerebral ischemia/reperfusion injury in rats. CONCLUSION Our results suggest that ginsenoside Rg1 can ameliorate cerebral ischemia-reperfusion injury by modulating Pink1/ Parkin-mediated mitochondrial autophagy in microglia and inhibiting microglial NLRP3 activation.
Collapse
Affiliation(s)
- Changbai Sui
- Department of Neurology, Yantaishan Hospital, Yantai 264001, China
| | - Ying Liu
- Department of Neurology, Yantaishan Hospital, Yantai 264001, China
| | - Jun Jiang
- Key Laboratory of Genetics Research and Evaluation of the National Drug Administration, Shandong Institute for Food and Drug Control, Shandong, Jinan 250033, China
| | - Jianhua Tang
- Department of Neurology, Yantaishan Hospital, Yantai 264001, China
| | - Ling Yu
- Department of Neurology, Yantaishan Hospital, Yantai 264001, China
| | - Guoying Lv
- Department of Anesthesiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China.
| |
Collapse
|
8
|
Lei SY, Qu Y, Yang YQ, Liu JC, Zhang YF, Zhou SY, He QY, Jin H, Yang Y, Guo ZN. Cellular senescence: A novel therapeutic target for central nervous system diseases. Biomed Pharmacother 2024; 179:117311. [PMID: 39182322 DOI: 10.1016/j.biopha.2024.117311] [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: 05/27/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
The underlying mechanisms of diseases affecting the central nervous system (CNS) remain unclear, limiting the development of effective therapeutic strategies. Remarkably, cellular senescence, a biological phenomenon observed in cultured fibroblasts in vitro, is a crucial intrinsic mechanism that influences homeostasis of the brain microenvironment and contributes to the onset and progression of CNS diseases. Cellular senescence has been observed in disease models established in vitro and in vivo and in bodily fluids or tissue components from patients with CNS diseases. These findings highlight cellular senescence as a promising target for preventing and treating CNS diseases. Consequently, emerging novel therapies targeting senescent cells have exhibited promising therapeutic effects in preclinical and clinical studies on aging-related diseases. These innovative therapies can potentially delay brain cell loss and functional changes, improve the prognosis of CNS diseases, and provide alternative treatments for patients. In this study, we examined the relevant advancements in this field, particularly focusing on the targeting of senescent cells in the brain for the treatment of chronic neurodegenerative diseases (e.g., Alzheimer's disease, Parkinson's disease, and multiple sclerosis) and acute neurotraumatic insults (e.g., ischemic stroke, spinal cord injury, and traumatic brain injury).
Collapse
Affiliation(s)
- Shuang-Yin Lei
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Yu-Qian Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Jia-Cheng Liu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Yi-Fei Zhang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Sheng-Yu Zhou
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Hang Jin
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China; Neuroscience Research Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
9
|
Peng G, Huang Y, Xie G, Tang J. Exploring Copper's role in stroke: progress and treatment approaches. Front Pharmacol 2024; 15:1409317. [PMID: 39391696 PMCID: PMC11464477 DOI: 10.3389/fphar.2024.1409317] [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: 03/29/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Copper is an important mineral, and moderate copper is required to maintain physiological processes in nervous system including cerebral ischemia/reperfusion (I/R) injury. Over the past few decades, copper induced cell death, named cuprotosis, has attracted increasing attention. Several lines of evidence have confirmed cuprotosis exerts pivotal role in diverse of pathological processes, such as cancer, neurodegenerative diseases, and I/R injury. Therefore, an in-depth understanding of the interaction mechanism between copper-mediated cell death and I/R injury may reveal the significant alterations about cellular copper-mediated homeostasis in physiological and pathophysiological conditions, as well as therapeutic strategies deciphering copper-induced cell death in cerebral I/R injury.
Collapse
Affiliation(s)
- Gang Peng
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan, China
| | - Yongpan Huang
- School of Medicine, Changsha Social Work College, Changsha, Hunan, China
| | - Guangdi Xie
- Department of Neurology, Huitong People’s Hospital, Huitong, Hunan, China
| | - Jiayu Tang
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
10
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 DOI: 10.1038/s41392-024-01933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
11
|
Xu W, Guo Y, Zhao L, Fu R, Qin X, Zhang Y, Cheng X, Xu S. The Aging Immune System: A Critical Attack on Ischemic Stroke. Mol Neurobiol 2024:10.1007/s12035-024-04464-2. [PMID: 39271626 DOI: 10.1007/s12035-024-04464-2] [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: 11/03/2023] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
Ischemic stroke caused by cerebrovascular embolism is an age-related disease with high rates of disability and mortality. Although the mechanisms of immune and inflammatory development after stroke have been of great interest, most studies have neglected the critical and unavoidable factor of age. As the global aging trend intensifies, the number of stroke patients is constantly increasing, emphasizing the urgency of finding effective measures to address the needs of elderly stroke patients. The concept of "immunosenescence" appears to explain the worse stroke outcomes in older individuals. Immune remodeling due to aging involves dynamic changes at all levels of the immune system, and the overall consequences of central (brain-resident) and peripheral (non-brain-resident) immune cells in stroke vary according to the age of the individual. Lastly, the review outlines recent strategies aimed at immunosenescence to improve stroke prognosis.
Collapse
Affiliation(s)
- Wenzhe Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuying Guo
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Linna Zhao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoli Qin
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xueqi Cheng
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
12
|
Lei Y, Meng J, Shi H, Shi C, Li C, Yang Z, Zhang W, Zuo D, Wang F, Wang M. Mannan-binding lectin inhibits oxidative stress-induced senescence via the NAD+/Sirt1 pathway. Int Immunopharmacol 2024; 137:112468. [PMID: 38906004 DOI: 10.1016/j.intimp.2024.112468] [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: 02/28/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024]
Abstract
Prolonged or excessive oxidative stress can lead to premature cellular and body aging. Mannan-binding lectin (MBL) is synthesized by the liver and plays an important role in innate immunity, anti-inflammation, and anti-oxidation, and has a positive impact on health and longevity. To date, few studies investigated the role of MBL in attenuating oxidative stress-induced senescence. In this study, we evaluated the role of MBL in oxidative stress-induced premature aging and explored its underlying mechanism in C57BL/6 mice and mouse embryonic fibroblasts (NIH/3T3). First, we established an oxidative premature senescence model induced by D-galactose in C57BL/6 mice. We found that MBL-deficient mice had a marked aging-like appearance, reduced learning and spatial exploration abilities, severe liver pathological damage, and significantly upregulated expression of Senescence-associated proteins (p53 and p21), inflammatory kinesins (IL-1β and IL-6), and the senescence β-galactosidase (SA-β-Gal) positive rate as compared with WT mice. In the H2O2-induced oxidative senescence model of NIH/3T3 cells, consistent results were obtained after MBL intervention. In addition, MBL effectively inhibited G1 phase arrest, ROS levels, DNA damage, and mitochondrial dysfunction in premature senescent cells. Mechanistically, we found that oxidative stress inhibited the nicotinamide adenine dinucleotide (NAD+)/ silent information regulator 1 (Sirt1) signaling pathway, while MBL activated the NAD+/Sirt1 signaling pathway inhibited by oxidative stress. In addition, MBL could activate the NAD+/Sirt1 pathway by upregulating NAMPT, which in turn inhibited p38 phosphorylation by activating the NAD+/Sirt1 pathway. In conclusion, MBL inhibits oxidative aging, which may facilitate the development of therapeutics to delay oxidative aging.
Collapse
Affiliation(s)
- Yiming Lei
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Jie Meng
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Haiqiang Shi
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| | - Chenchen Shi
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Chao Li
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ziyi Yang
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Wei Zhang
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang 453003, China
| | - Daming Zuo
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Fanping Wang
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Mingyong Wang
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, China; School of Medical Technology, Shangqiu Medical College, Shangqiu 476100, China.
| |
Collapse
|
13
|
Zhou X, Luo X. Shionone relieves oxygen-glucose deprivation/reoxygenation induced SH-SY5Y cells injury by inhibiting the p38 MAPK/NF-κB pathway. J Cardiothorac Surg 2024; 19:435. [PMID: 38997740 PMCID: PMC11241947 DOI: 10.1186/s13019-024-02938-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/16/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (I/R) can affect patient outcomes and can even be life-threatening. This study aimed to explore the role of Shionone in cerebral I/R and reveal its mechanism of action through the cerebral I/R in vitro model. METHODS SH-SY5Y cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to induce cerebral I/R in vitro model. SH-SY5Y cells were treated with different concentrations of Shionone. Cell counting kit-8 and flow cytometry assays were used to detect cell viability and apoptosis levels. The levels of superoxide dismutase, catalase, and malondialdehyde were determined using their corresponding kits to examine the level of oxidative stress. The inflammation response was detected by IL-6, IL-1β, and TNF-α levels, using enzyme-linked-immunosorbent-assay. RT-qPCR was performed to measure the mRNA levels of p38 and NF-κB. Western blotting was used to quantify the apoptosis-related proteins and p38MAPK/NF-κB signaling pathway proteins. RESULTS Shionone exhibited no toxic effects on SH-SY5Y cells. Shionone inhibited OGD/R-induced cell apoptosis, improved the inflammatory response caused by OGD/R, and reduced the level of oxidative stress in cells. Western blot assay results showed that Shionone alleviated OGD/R-induced injury by inhibiting the activity of the p38 MAPK/NF-κB signaling pathway. The p38/MAPK agonist P79350 reversed the beneficial effects of Shionone. CONCLUSION Shionone alleviates cerebral I/R and may thus be a novel therapeutic strategy for treating cerebral I/R.
Collapse
Affiliation(s)
- Xiaoli Zhou
- Department of Neurology, The Affiliated Hospital of Hubei University of Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Xueting Luo
- Department of Cardiovascular Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, No. 856 Luoyu Road, Hongshan District, Wuhan, 430061, China.
| |
Collapse
|
14
|
Qi M, Su X, Li Z, Huang H, Wang J, Lin N, Kong X. Bibliometric analysis of research progress on tetramethylpyrazine and its effects on ischemia-reperfusion injury. Pharmacol Ther 2024; 259:108656. [PMID: 38735486 DOI: 10.1016/j.pharmthera.2024.108656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/22/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
In recent decades, natural products have attracted worldwide attention and become one of the most important resources for pharmacological industries and medical sciences to identify novel drug candidates for disease treatment. Tetramethylpyrazine (TMP) is an alkaloid extracted from Ligusticum chuanxiong Hort., which has shown great therapeutic potential in cardiovascular and cerebrovascular diseases, liver and renal injury, as well as cancer. In this review, we analyzed 1270 papers published on the Web of Science Core Collection from 2002 to 2022 and found that TMP exerted significant protective effects on ischemia-reperfusion (I/R) injury that is the cause of pathological damages in a variety of conditions, such as ischemic stroke, myocardial infarction, acute kidney injury, and liver transplantation. TMP is limited in clinical applications to some extent due to its rapid metabolism, a short biological half-life and poor bioavailability. Obviously, the structural modification, administration methods and dosage forms of TMP need to be further investigated in order to improve its bioavailability. This review summarizes the clinical applications of TMP, elucidates its potential mechanisms in protecting I/R injury, provides strategies to improve bioavailability, which presents a comprehensive understanding of the important compound. Hopefully, the information and knowledge from this review can help researchers and physicians to better improve the applications of TMP in the clinic.
Collapse
Affiliation(s)
- Mingzhu Qi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaohui Su
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhuohang Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Helan Huang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jingbo Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Na Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiangying Kong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
15
|
Chen Q, Wu M, Tang Q, Yan P, Zhu L. Age-Related Alterations in Immune Function and Inflammation: Focus on Ischemic Stroke. Aging Dis 2024; 15:1046-1074. [PMID: 37728582 PMCID: PMC11081165 DOI: 10.14336/ad.2023.0721-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/21/2023] [Indexed: 09/21/2023] Open
Abstract
The aging of the global population poses significant scientific challenges. Moreover, the biological process of aging is the most significant risk factor for most chronic illnesses; therefore, understanding the molecular and cellular mechanisms underlying these aging-related challenges is crucial for extending the healthy lifespan of older individuals. Preventing brain aging remains a priority public health goal, and integrative and comprehensive aging analyses have revealed that immunosenescence is a potential cause of age-related brain damage and disease (e.g., stroke). Importantly, the neuroinflammatory and immune systems present two-way contact and thus can affect each other. Emerging evidence supports the numerous effects of immunosenescence- and inflammation-mediated immunity in neurologically injured brains. In this study, we briefly outline how aging alters the pathophysiology and transcriptional amplitude in patients who experienced stroke and then discuss how the immune system and its cellular components and molecular mechanisms are affected by age after stroke. Finally, we highlight emerging interventions with the potential to slow down or reduce aging and prevent stroke onset.
Collapse
Affiliation(s)
- Qiuxin Chen
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Minmin Wu
- Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Qiang Tang
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Peiyu Yan
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Luwen Zhu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150000, China
| |
Collapse
|
16
|
Badran M, Puech C, Khalyfa A, Cortese R, Cataldo K, Qiao Z, Gozal D. Senolytic-facilitated Reversal of End-Organ Dysfunction in a Murine Model of Obstructive Sleep Apnea. Am J Respir Crit Care Med 2024; 209:1001-1012. [PMID: 38113165 PMCID: PMC11531216 DOI: 10.1164/rccm.202306-1101oc] [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: 06/26/2023] [Accepted: 12/19/2023] [Indexed: 12/21/2023] Open
Abstract
Rationale: Obstructive sleep apnea (OSA) is a highly prevalent condition that is associated with accelerated biological aging and multiple end-organ morbidities. Current treatments, such as continuous positive airway pressure (CPAP), have shown limited cognitive, metabolic, and cardiovascular beneficial outcomes despite adherence. Thus, adjunct therapies aiming to reduce OSA burden, such as senolytics, could improve OSA outcomes.Objectives: To assess if targeting senescence in addition to partial normoxia mimicking "good" CPAP adherence can improve physiological outcomes in mice exposed to chronic intermittent hypoxia.Methods: We compared the effects of 6 weeks of therapy with either partial normoxic recovery alone or combined with the senolytic navitoclax after 16 weeks of intermittent hypoxia exposures, a hallmark of OSA, on multiphenotypic cardiometabolic and neurocognitive parameters.Measurements and Main Results: Our findings indicate that only when combined with navitoclax, partial normoxic recovery significantly improved sleepiness (sleep in the dark phase: 34% ± 4% vs. 26% ± 3%; P < 0.01), cognition (preference score: 51% ± 19% vs. 70% ± 11%; P = 0.048), coronary artery function (response to acetylcholine [vasodilation]: 56% ± 13% vs. 72% ± 10%; P < 0.001), glucose, and lipid metabolism and reduced intestinal permeability and senescence in multiple organs.Conclusions: These findings indicate that the reversibility of end-organ morbidities induced by OSA is not only contingent on restoration of normal oxygenation patterns but can be further enhanced by targeting other OSA-mediated detrimental cellular processes, such as accelerated senescence.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri; and
| | - Clementine Puech
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri; and
| | - Abdelnaby Khalyfa
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri; and
| | - Rene Cortese
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri; and
| | - Kylie Cataldo
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri; and
| | - Zhuanhong Qiao
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri; and
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri; and
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| |
Collapse
|
17
|
Kumar M, Yan P, Kuchel GA, Xu M. Cellular Senescence as a Targetable Risk Factor for Cardiovascular Diseases: Therapeutic Implications: JACC Family Series. JACC Basic Transl Sci 2024; 9:522-534. [PMID: 38680957 PMCID: PMC11055207 DOI: 10.1016/j.jacbts.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 05/01/2024]
Abstract
The prevalence of cardiovascular diseases markedly rises with age. Cellular senescence, a hallmark of aging, is characterized by irreversible cell cycle arrest and the manifestation of a senescence-associated secretory phenotype, which has emerged as a significant contributor to aging, mortality, and a spectrum of chronic ailments. An increasing body of preclinical and clinical research has established connections between senescence, senescence-associated secretory phenotype, and age-related cardiac and vascular pathologies. This review comprehensively outlines studies delving into the detrimental impact of senescence on various cardiovascular diseases, encompassing systemic atherosclerosis (including coronary artery disease, stroke, and peripheral arterial disease), as well as conditions such as hypertension, congestive heart failure, arrhythmias, and valvular heart diseases. In addition, we have preclinical studies demonstrating the beneficial effects of senolytics-a class of drugs designed to eliminate senescent cells selectively across diverse cardiovascular disease scenarios. Finally, we address knowledge gaps on the influence of senescence on cardiovascular systems and discuss the future trajectory of strategies targeting senescence for cardiovascular diseases.
Collapse
Affiliation(s)
- Manish Kumar
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Division of Critical Care Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Pengyi Yan
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - George A. Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Ming Xu
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
18
|
Li J, Xie F, Ma X. Advances in nanomedicines: a promising therapeutic strategy for ischemic cerebral stroke treatment. Nanomedicine (Lond) 2024; 19:811-835. [PMID: 38445614 DOI: 10.2217/nnm-2023-0266] [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: 03/07/2024] Open
Abstract
Ischemic stroke, prevalent among the elderly, necessitates attention to reperfusion injury post treatment. Limited drug access to the brain, owing to the blood-brain barrier, restricts clinical applications. Identifying efficient drug carriers capable of penetrating this barrier is crucial. Blood-brain barrier transporters play a vital role in nutrient transport to the brain. Recently, nanoparticles emerged as drug carriers, enhancing drug permeability via surface-modified ligands. This article introduces the blood-brain barrier structure, elucidates reperfusion injury pathogenesis, compiles ischemic stroke treatment drugs, explores nanomaterials for drug encapsulation and emphasizes their advantages over conventional drugs. Utilizing nanoparticles as drug-delivery systems offers targeting and efficiency benefits absent in traditional drugs. The prospects for nanomedicine in stroke treatment are promising.
Collapse
Affiliation(s)
- Jun Li
- Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, PR China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, PR China
| | - Fei Xie
- Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, PR China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, PR China
| | - Xuemei Ma
- Faculty of Environment & Life, Beijing University of Technology, Beijing, 100124, PR China
- Beijing Molecular Hydrogen Research Center, Beijing, 100124, PR China
| |
Collapse
|
19
|
Vercalsteren E, Karampatsi D, Buizza C, Nyström T, Klein T, Paul G, Patrone C, Darsalia V. The SGLT2 inhibitor Empagliflozin promotes post-stroke functional recovery in diabetic mice. Cardiovasc Diabetol 2024; 23:88. [PMID: 38424560 PMCID: PMC10905950 DOI: 10.1186/s12933-024-02174-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
Type-2 diabetes (T2D) worsens stroke recovery, amplifying post-stroke disabilities. Currently, there are no therapies targeting this important clinical problem. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) are potent anti-diabetic drugs that also efficiently reduce cardiovascular death and heart failure. In addition, SGLT2i facilitate several processes implicated in stroke recovery. However, the potential efficacy of SGLT2i to improve stroke recovery in T2D has not been investigated. Therefore, we determined whether a post-stroke intervention with the SGLT2i Empagliflozin could improve stroke recovery in T2D mice. T2D was induced in C57BL6J mice by 8 months of high-fat diet feeding. Hereafter, animals were subjected to transient middle cerebral artery occlusion and treated with vehicle or the SGLTi Empagliflozin (10 mg/kg/day) starting from 3 days after stroke. A similar study in non diabetic mice was also conducted. Stroke recovery was assessed using the forepaw grip strength test. To identify potential mechanisms involved in the Empagliflozin-mediated effects, several metabolic parameters were assessed. Additionally, neuronal survival, neuroinflammation, neurogenesis and cerebral vascularization were analyzed using immunohistochemistry/quantitative microscopy. Empagliflozin significantly improved stroke recovery in T2D but not in non-diabetic mice. Improvement of functional recovery was associated with lowered glycemia, increased serum levels of fibroblast growth factor-21 (FGF-21), and the normalization of T2D-induced aberration of parenchymal pericyte density. The global T2D-epidemic and the fact that T2D is a major risk factor for stroke are drastically increasing the number of people in need of efficacious therapies to improve stroke recovery. Our data provide a strong incentive for the potential use of SGLT2i for the treatment of post-stroke sequelae in T2D.
Collapse
Affiliation(s)
- Ellen Vercalsteren
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden.
| | - Dimitra Karampatsi
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Carolina Buizza
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Thomas Nyström
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Cesare Patrone
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden.
| | - Vladimer Darsalia
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden.
| |
Collapse
|
20
|
Melo Dos Santos LS, Trombetta-Lima M, Eggen B, Demaria M. Cellular senescence in brain aging and neurodegeneration. Ageing Res Rev 2024; 93:102141. [PMID: 38030088 DOI: 10.1016/j.arr.2023.102141] [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: 09/28/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Cellular senescence is a state of terminal cell cycle arrest associated with various macromolecular changes and a hypersecretory phenotype. In the brain, senescent cells naturally accumulate during aging and at sites of age-related pathologies. Here, we discuss the recent advances in understanding the accumulation of senescent cells in brain aging and disorders. Here we highlight the phenotypical heterogeneity of different senescent brain cell types, highlighting the potential importance of subtype-specific features for physiology and pathology. We provide a comprehensive overview of various senescent cell types in naturally occurring aging and the most common neurodegenerative disorders. Finally, we critically discuss the potential of adapting senotherapeutics to improve brain health and reduce pathological progression, addressing limitations and future directions for application and development.
Collapse
Affiliation(s)
- L S Melo Dos Santos
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA, Groningen, the Netherlands; School of Sciences, Health and Life, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Avenue, 6681, 90619-900 Porto Alegre, Brazil
| | - M Trombetta-Lima
- Department of Biomedical Sciences of Cells and Systems, section Molecular Neurobiology, University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA Groningen, the Netherlands; Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusiglaan 1, 9713AV Groningen, the Netherlands
| | - Bjl Eggen
- Department of Biomedical Sciences of Cells and Systems, section Molecular Neurobiology, University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA Groningen, the Netherlands
| | - M Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9715RA, Groningen, the Netherlands.
| |
Collapse
|
21
|
Hui W, Song T, Yu L, Chen X. The Binding of HSPA8 and Mitochondrial ALDH2 Mediates Oxygen-Glucose Deprivation-Induced Fibroblast Senescence. Antioxidants (Basel) 2023; 13:42. [PMID: 38247467 PMCID: PMC10812545 DOI: 10.3390/antiox13010042] [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: 11/16/2023] [Revised: 12/17/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Cellular senescence refers to the permanent and irreversible cessation of the cell cycle. Recently, it has gained significant interest as a promising target for preventing cardiovascular diseases. Aldehyde dehydrogenase 2 (ALDH2) is a mitochondrial enzyme that has been closely linked with an increased risk of cardiovascular diseases. In this study, bioinformatics analysis revealed that the signaling pathway for fibroblast senescence is significantly activated in mice after myocardial infarction (MI), and that ALDH2 might be a crucial molecule responsible for inducing this change. Therefore, we created an NIH3T3 fibroblast cell line oxygen-glucose deprivation (OGD) model to replicate the conditions of MI in vitro. We further revealed that decreased ALDH2 enzyme activity is a critical factor that affects fibroblast senescence after OGD, and the activation of ALDH2 can improve the mitochondrial damage caused by OGD. We identified Heat Shock 70-kDa Protein 8 (HSPA8) as an interacting protein of ALDH2 through co-immunoprecipitation (Co-IP) and mass spectrometry (MS) detection. Subsequently, our studies showed that HSPA8 translocates to the mitochondria after OGD, potentially binding to ALDH2 and inhibiting its enzyme activity. By transfecting siRNA to inhibit HSPA8 expression in cells, it was found that ALDH2 enzyme activity can be significantly increased, and the senescence characteristics induced by OGD in NIH3T3 cells can be improved. In conclusion, the data from this study suggest that HSPA8, in conjunction with ALDH2, could regulate fibroblast senescence after oxygen-glucose deprivation, providing a new direction and foundation for effectively intervening in fibroblast senescence after myocardial infarction.
Collapse
Affiliation(s)
- Wenting Hui
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China;
| | - Tongtong Song
- Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130012, China;
| | - Ling Yu
- Department of Pharmacy, The Second Hospital of Jilin University, Changchun 130022, China;
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130012, China;
| |
Collapse
|
22
|
Liu H, Shen L, Sun Z, Wu W, Xu M. Downregulated PGK1 attenuates cerebral ischemia-reperfusion injury by reversing neuroinflammation and oxidative stress through the Nrf2/ARE pathway. Neuroscience 2023:S0306-4522(23)00239-7. [PMID: 37295596 DOI: 10.1016/j.neuroscience.2023.05.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
Understanding the role and mechanism of astrocytes in inflammation and oxidative response is crucial for developing therapeutic strategies to reduce inflammation and oxidative injury in cerebral ischemia-reperfusion injury (CIRI). In this study, we investigated the regulatory effects of phosphoglycerate kinase 1 (PGK1) on inflammation and oxidative response after CIRI in male adult Sprague-Dawley (SD) rats and using primary astrocytes obtained from neonatal SD rats, and explored its related mechanisms. We established a rat model of middle cerebral artery occlusion-reperfusion (MCAO/R) by suture occlusion, and an oxygen-glucose deprivation/reoxygenation model of astrocytes using oxygen-free, glucose-free, and serum-free cultures. AAV8-PGK1-GFP was injected into the left ventricle 24 h before modeling. Real-time quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, co-immunoprecipitation (CoIP) assay, fluorescence in situ hybridization (FISH), and western blotting were used to elucidate the in-depth mechanisms of PGK1 in CIRI. PGK1 overexpression significantly exacerbated neurological deficits, increased cerebral infarct volume, and aggravated nerve cell injury in rats after MCAO/R. Using FISH and CoIP assays, we verified the localization of PGK1 and Nrf2 in primary astrocytes. Further rescue experiments showed that Nrf2 knockdown eliminated the protective effect of CBR-470-1 (a PGK1 inhibitor) on CIRI. Lastly, we confirmed that PGK1 aggravates CIRI by inhibiting the Nrf2/ARE pathway. In conclusion, our findings suggest that inhibiting PGK1 attenuates CIRI by reducing the release of inflammatory and oxidative factors from astrocytes by activating the Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Hua Liu
- Department of Neurosurgery, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, 215300, China
| | - Likui Shen
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, 215028, China
| | - Zezhi Sun
- Department of Neurosurgery, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, 215300, China
| | - Wenxi Wu
- Department of Neurosurgery, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, 215300, China
| | - Min Xu
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, 215300, China.
| |
Collapse
|
23
|
Knopp RC, Erickson MA, Rhea EM, Reed MJ, Banks WA. Cellular senescence and the blood-brain barrier: Implications for aging and age-related diseases. Exp Biol Med (Maywood) 2023; 248:399-411. [PMID: 37012666 PMCID: PMC10281623 DOI: 10.1177/15353702231157917] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
The blood-brain barrier (BBB) is a critical physiochemical interface that regulates communication between the brain and blood. It is comprised of brain endothelial cells which regulate the BBB's barrier and interface properties and is surrounded by supportive brain cell types including pericytes and astrocytes. Recent reports have suggested that the BBB undergoes dysfunction during normative aging and in disease. In this review, we consider the effect of cellular senescence, one of the nine hallmarks of aging, on the BBB. We first characterize known normative age-related changes at the BBB, and then evaluate changes in neurodegenerative diseases, with an emphasis on if/how cellular senescence is influencing these changes. We then discuss what insight has been gained from in vitro and in vivo studies of cellular senescence at the BBB. Finally, we evaluate mechanisms by which cellular senescence in peripheral pathologies can indirectly or directly affect BBB function.
Collapse
Affiliation(s)
- Rachel C Knopp
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Michelle A Erickson
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - May J Reed
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| |
Collapse
|
24
|
Vitexin Improves Cerebral ischemia‑reperfusion Injury by Attenuating Oxidative Injury and Ferroptosis via Keap1/Nrf2/HO-1signaling. Neurochem Res 2023; 48:980-995. [PMID: 36435955 DOI: 10.1007/s11064-022-03829-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 11/14/2022] [Indexed: 11/28/2022]
Abstract
Cerebral ischemia/reperfusion involves multiple pathological processes and ferroptosis played a crucial role in the disease progression. Nevertheless, whether Vitexin could ameliorate ischemia/reperfusion injury via meditate the ferroptosis still remains unknown. In this study, we established the oxygen-glucose deprivation and reoxygenation (OGD/R) neuron cell and middle cerebral artery occlusion/reperfusion (MCAO/R) rat model. The cell viability, cell apoptosis and reactive oxygen species (ROS) levels were tested by CCK-8 assay and Flow cytometry, respectively. Hematoxylin-eosin staining, TTC, TEM, immunofluorescence analysis and western blot were used to investigate the effects of Vitexin. The results demonstrated that Vitexin could enhanced the cell viability and decreased the cell apoptosis in OGD/R cell model. Meanwhile, incubation with Vitexin maintained the neuroprotective effects in OGD/R induced generation of lipid ROS and neuronal cell ferroptosis via regulated the expressions of Keap1/Nrf2/HO-1 relative protein levels. Moreover, treatment with Vitexin reversed brain infracted volume, the normal histopathology and mitochondrial function in MCAO/R rat model. Vitexin significantly decreased the Nrf2 transfer ration from nuclear to cytosol and regulated the expression of Keap1/Nrf2/HO-1 signaling both in vitro and in vivo. Nevertheless, the protective effects of Vitexin were blocked with the Nrf2 inhibitor ML385. Vitexin could protect the neuron cell and brain related with the Keap1/Nrf2/HO-1 signaling pathway. Vitexin was a useful candidate for stroke therapy and our research may provide an attractive therapeutic target for the treatment of stroke.
Collapse
|
25
|
Zhao J, Ma M, Li L, Fang G. Oxysophoridine protects against cerebral ischemia/reperfusion injury via inhibition of TLR4/p38MAPK‑mediated ferroptosis. Mol Med Rep 2023; 27:44. [PMID: 36601753 PMCID: PMC9887505 DOI: 10.3892/mmr.2023.12931] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/08/2022] [Indexed: 01/05/2023] Open
Abstract
Oxysophoridine (OSR) is an alkaloid extracted from Sophora alopecuroides L. and exerts beneficial effects in cerebral ischemia/reperfusion (I/R) injury. However, the molecular mechanism underlying the regulatory effects of OSR in cerebral I/R injury remains unclear. In the present study, a cerebral I/R injury rat model was established by occlusion of the right middle cerebral artery. Hematoxylin and eosin and triphenyltetrazolium chloride staining were performed to assess histopathological changes and the extent of cerebral injury to the brain. A Cell Counting Kit‑8 and TUNEL assay and western blotting were performed to assess cell viability and apoptosis. Ferroptosis and oxidative stress were evaluated based on ATP and Fe2+ levels and DCFH‑DA staining. The protein expression levels of inflammatory factors were assessed using ELISA. The protein expression levels of members of the toll‑like receptor (TLR)4/p38MAPK signaling pathway were evaluated using immunofluorescence staining and western blotting. The results demonstrated that OSR decreased brain injury and neuronal apoptosis in the hippocampus in I/R‑induced rats. OSR inhibited reactive oxygen species (ROS) production, decreased levels of ATP, Fe2+ and acyl‑CoA synthetase long‑chain family member 4 (ACSL4) and transferrin 1 protein and increased the protein expression levels of ferritin 1 and glutathione peroxidase 4. Furthermore, OSR blocked TLR4/p38MAPK signaling in brain tissue in the I/R‑induced rat. In vitro experiments demonstrated that TLR4 overexpression induced generation of ROS, ATP and Fe2+, which promoted the expression of ferroptosis‑associated proteins in hippocampal HT22 neuronal cells. The ferroptosis inducer erastin decreased the effects of OSR on oxygen‑glucose deprivation/reoxygenation (OGD/R)‑induced cell viability, oxidative stress and inflammatory response. Together, the results demonstrated that OSR alleviated cerebral I/R injury via inhibition of TLR4/p38MAPK‑mediated ferroptosis.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Neurology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China,Correspondence to: Dr Jing Zhao, Department of Neurology, Hangzhou Red Cross Hospital, 208 Eastern Ring Road, Hangzhou, Zhejiang 310003, P.R. China, E-mail:
| | - Mingming Ma
- Department of Neurology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Lei Li
- Department of Neurology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Gaoli Fang
- Department of Neurology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
26
|
Liu M, Mu J, Gong W, Zhang K, Yuan M, Song Y, Li B, Jin N, Zhang W, Zhang D. In Vitro Diagnosis and Visualization of Cerebral Ischemia/Reperfusion Injury in Rats and Protective Effects of Ferulic Acid by Raman Biospectroscopy and Machine Learning. ACS Chem Neurosci 2023; 14:159-169. [PMID: 36516359 DOI: 10.1021/acschemneuro.2c00612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is a major cause of mortality with complicated pathophysiological mechanisms, and hematoxylin and eosin (HE) staining is a histochemical diagnosis technique heavily relying on subjective observation. In this study, we developed a noninvasive assay using Raman spectroscopy for in vitro diagnosis and visualization of cerebral ischemia/reperfusion injury and protective effects of ferulic acid. By establishing a middle cerebral artery occlusion (MCAO) model in Sprague-Dawley male rats, we found effective interventions by ferulic acid using the neurological function score and HE staining. Raman spectra of neuronal and neuroglial cells exhibited significant intensity changes of protein, nucleotide, lipid, and carbohydrate at 780, 814, 1002, 1012, 1176, 1224, 1402, 1520, 1586, 1614, and 1752 cm-1. Cluster vector analysis highlighted the alterations at 1002, 1080, 1298, 1430, 1478, 1508, 1586, and 1676 cm-1. To evaluate the levels of neuron injury and intervention performance, a random forest model was developed on Raman spectral data and achieved satisfactory accuracy (0.9846), sensitivity (0.9679-0.9932), and specificity (0.9945-0.9989), ranking peaks around 1002 cm-1 as key fingerprint for classification. Spectral phenylalanine-to-tryptophan ratio was the biomarker to visualize neuronal injury and intervention performance of ferulic acid with a resolution of 1 μm. Our results unravel the biochemical changes in neuronal cells with cerebral ischemia/reperfusion injury and ferulic acid treatment, and prove Raman spectroscopy coupled with machine learning as a power tool to classify neuron viability and evaluate the intervention performance in pharmacological research.
Collapse
Affiliation(s)
- Mingying Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou310053, P. R. China
| | - Ju Mu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou310053, P. R. China
| | - Wan Gong
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou310053, P. R. China
| | - Kena Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou310053, P. R. China
| | - Maoyun Yuan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou310053, P. R. China
| | - Yizhi Song
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, P. R. China
| | - Bei Li
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun130033, P. R. China
| | - Naifu Jin
- College of Water Sciences, Beijing Normal University, Beijing100875, P. R. China
| | - Wenjing Zhang
- Key Laboratory of Groundwater Resources and Environment, Ministry of Education, Changchun130021, P. R. China.,College of New Energy and Environment, Jilin University, Changchun130021, P. R. China
| | - Dayi Zhang
- Key Laboratory of Groundwater Resources and Environment, Ministry of Education, Changchun130021, P. R. China.,College of New Energy and Environment, Jilin University, Changchun130021, P. R. China
| |
Collapse
|
27
|
Luo H, Guo H, Zhou Y, Fang R, Zhang W, Mei Z. Neutrophil Extracellular Traps in Cerebral Ischemia/Reperfusion Injury: Friend and Foe. Curr Neuropharmacol 2023; 21:2079-2096. [PMID: 36892020 PMCID: PMC10556361 DOI: 10.2174/1570159x21666230308090351] [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: 10/25/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 03/10/2023] Open
Abstract
Cerebral ischemic injury, one of the leading causes of morbidity and mortality worldwide, triggers various central nervous system (CNS) diseases, including acute ischemic stroke (AIS) and chronic ischemia-induced Alzheimer's disease (AD). Currently, targeted therapies are urgently needed to address neurological disorders caused by cerebral ischemia/reperfusion injury (CI/RI), and the emergence of neutrophil extracellular traps (NETs) may be able to relieve the pressure. Neutrophils are precursors to brain injury following ischemic stroke and exert complicated functions. NETs extracellularly release reticular complexes of neutrophils, i.e., double-stranded DNA (dsDNA), histones, and granulins. Paradoxically, NETs play a dual role, friend and foe, under different conditions, for example, physiological circumstances, infection, neurodegeneration, and ischemia/reperfusion. Increasing evidence indicates that NETs exert anti-inflammatory effects by degrading cytokines and chemokines through protease at a relatively stable and moderate level under physiological conditions, while excessive amounts of NETs release (NETosis) irritated by CI/RI exacerbate the inflammatory response and aggravate thrombosis, disrupt the blood-brain barrier (BBB), and initiates sequential neuron injury and tissue damage. This review provides a comprehensive overview of the machinery of NETs formation and the role of an abnormal cascade of NETs in CI/RI, as well as other ischemia-induced neurological diseases. Herein, we highlight the potential of NETs as a therapeutic target against ischemic stroke that may inspire translational research and innovative clinical approaches.
Collapse
Affiliation(s)
- Haoyue Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Hanjing Guo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Yue Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Rui Fang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| |
Collapse
|
28
|
Wu Y, Fan X, Chen S, Deng L, Jiang L, Yang S, Dong Z. Geraniol-Mediated Suppression of Endoplasmic Reticulum Stress Protects against Cerebral Ischemia-Reperfusion Injury via the PERK-ATF4-CHOP Pathway. Int J Mol Sci 2022; 24:ijms24010544. [PMID: 36613992 PMCID: PMC9820715 DOI: 10.3390/ijms24010544] [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] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 12/31/2022] Open
Abstract
Endoplasmic reticulum (ER) stress plays an important role in cerebral ischemia-reperfusion injury (CIRI). Geraniol has antioxidant, antibacterial, and anti-inflammatory activities. Studies have shown that geraniol has a protective effect against CIRI in rats, but the exact mechanism is unclear. Purpose: The aim of this study was to investigate the protective mechanism of geraniol against CIRI. We established a middle cerebral artery occlusion reperfusion model in rats and a PC12 cell oxygen-glucose deprivation/reoxygenation (OGD/R) model to observe the neuroprotective effects of geraniol. Neurological scoring, 2,3,5-triphenyltetrazolium chloride staining, and hematoxylin and eosin staining were used to evaluate the neuroprotective effects of geraniol against CIRI. ER-stress-related and apoptosis-related protein expression was detected via Western blotting and immunofluorescence. Apoptosis was also detected via TUNEL assays and flow cytometry. The fluorescent detection of intracellular calcium was achieved using fluorescent calcium-binding dyes, and transmission electron microscopy was used to assess the neuronal ultrastructure. Geraniol effectively attenuated cerebral infarction and pathological injury after CIRI, had a protective effect against CIRI, significantly reduced the expression of the ER-stress-related proteins P-PERK, ATF4, CHOP, and GRP78 and the pro-apoptotic protein BAX, increased the expression of the anti-apoptotic protein BCL-2, and reduced the occurrence of apoptosis. In the OGD/R model in PC12 cells, the protective effect of geraniol was the same as that in vivo. Our results suggest that geraniol has a protective effect against ischemic stroke by a mechanism possibly related to ER stress via the PERK-ATF4-CHOP pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhi Dong
- Correspondence: ; Tel.: +86-135-0839-3231
| |
Collapse
|
29
|
Re-purposing the pro-senescence properties of doxorubicin to introduce immunotherapy in breast cancer brain metastasis. Cell Rep Med 2022; 3:100821. [PMID: 36384097 PMCID: PMC9729880 DOI: 10.1016/j.xcrm.2022.100821] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 09/02/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022]
Abstract
An increasing number of breast cancer patients develop brain metastases (BM). Standard-of-care treatments are largely inefficient, and breast cancer brain metastasis (BCBM) patients are considered untreatable. Immunotherapies are not successfully employed in BCBM, in part because breast cancer is a "cold" tumor and also because the brain tissue has a unique immune landscape. Here, we generate and characterize immunocompetent models of BCBM derived from PyMT and Neu mammary tumors to test how harnessing the pro-senescence properties of doxorubicin can be used to prime the specific immune BCBM microenvironment. We reveal that BCBM senescent cells, induced by doxorubicin, trigger the recruitment of PD1-expressing T cells to the brain. Importantly, we demonstrate that induction of senescence with doxorubicin improves the efficacy of immunotherapy with anti-PD1 in BCBM in a CD8 T cell-dependent manner, thereby providing an optimized strategy to introduce immune-based treatments in this lethal disease. In addition, our BCBM models can be used for pre-clinical testing of other therapeutic strategies in the future.
Collapse
|
30
|
Wei Z, Xie Y, Wei M, Zhao H, Ren K, Feng Q, Xu Y. New insights in ferroptosis: Potential therapeutic targets for the treatment of ischemic stroke. Front Pharmacol 2022; 13:1020918. [PMID: 36425577 PMCID: PMC9679292 DOI: 10.3389/fphar.2022.1020918] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/26/2022] [Indexed: 10/22/2023] Open
Abstract
Stroke is a common disease in clinical practice, which seriously endangers people's physical and mental health. The neurovascular unit (NVU) plays a key role in the occurrence and development of ischemic stroke. Different from other classical types of cell death such as apoptosis, necrosis, autophagy, and pyroptosis, ferroptosis is an iron-dependent lipid peroxidation-driven new form of cell death. Interestingly, the function of NVU and stroke development can be regulated by activating or inhibiting ferroptosis. This review systematically describes the NVU in ischemic stroke, provides a comprehensive overview of the regulatory mechanisms and key regulators of ferroptosis, and uncovers the role of ferroptosis in the NVU and the progression of ischemic stroke. We further discuss the latest progress in the intervention of ferroptosis as a therapeutic target for ischemic stroke and summarize the research progress and regulatory mechanism of ferroptosis inhibitors on stroke. In conclusion, ferroptosis, as a new form of cell death, plays a key role in ischemic stroke and is expected to become a new therapeutic target for this disease.
Collapse
Affiliation(s)
- Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingze Wei
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Huijuan Zhao
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Kaidi Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Qi Feng
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Schwab N, Taskina D, Leung E, Innes BT, Bader GD, Hazrati LN. Neurons and glial cells acquire a senescent signature after repeated mild traumatic brain injury in a sex-dependent manner. Front Neurosci 2022; 16:1027116. [PMID: 36408415 PMCID: PMC9669743 DOI: 10.3389/fnins.2022.1027116] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/23/2022] [Indexed: 08/15/2023] Open
Abstract
Mild traumatic brain injury (mTBI) is an important public health issue, as it can lead to long-term neurological symptoms and risk of neurodegenerative disease. The pathophysiological mechanisms driving this remain unclear, and currently there are no effective therapies for mTBI. In this study on repeated mTBI (rmTBI), we have induced three mild closed-skull injuries or sham procedures, separated by 24 h, in C57BL/6 mice. We show that rmTBI mice have prolonged righting reflexes and astrogliosis, with neurological impairment in the Morris water maze (MWM) and the light dark test. Cortical and hippocampal tissue analysis revealed DNA damage in the form of double-strand breaks, oxidative damage, and R-loops, markers of cellular senescence including p16 and p21, and signaling mediated by the cGAS-STING pathway. This study identified novel sex differences after rmTBI in mice. Although these markers were all increased by rmTBI in both sexes, females had higher levels of DNA damage, lower levels of the senescence protein p16, and lower levels of cGAS-STING signaling proteins compared to their male counterparts. Single-cell RNA sequencing of the male rmTBI mouse brain revealed activation of the DNA damage response, evidence of cellular senescence, and pro-inflammatory markers reminiscent of the senescence-associated secretory phenotype (SASP) in neurons and glial cells. Cell-type specific changes were also present with evidence of brain immune activation, neurotransmission alterations in both excitatory and inhibitory neurons, and vascular dysfunction. Treatment of injured mice with the senolytic drug ABT263 significantly reduced markers of senescence only in males, but was not therapeutic in females. The reduction of senescence by ABT263 in male mice was accompanied by significantly improved performance in the MWM. This study provides compelling evidence that senescence contributes to brain dysfunction after rmTBI, but may do so in a sex-dependent manner.
Collapse
Affiliation(s)
- Nicole Schwab
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Daria Taskina
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Emily Leung
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Brendan T. Innes
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Gary D. Bader
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Lili-Naz Hazrati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
32
|
Cellular senescence in ischemia/reperfusion injury. Cell Death Dis 2022; 8:420. [PMID: 36253355 PMCID: PMC9576687 DOI: 10.1038/s41420-022-01205-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022]
Abstract
Ischemia/reperfusion (IR) injury, a main reason of mortality and morbidity worldwide, occurs in many organs and tissues. As a result of IR injury, senescent cells can accumulate in multiple organs. Increasing evidence shows that cellular senescence is the underlying mechanism that transforms an acute organ injury into a chronic one. Several recent studies suggest senescent cells can be targeted for the prevention or elimination of acute and chronic organ injury induced by IR. In this review, we concisely introduce the underlying mechanism and the pivotal role of premature senescence in the transition from acute to chronic IR injuries. Special focus is laid on recent advances in the mechanisms as well as on the basic and clinical research, targeting cellular senescence in multi-organ IR injuries. Besides, the potential directions in this field are discussed in the end. Together, the recent advances reviewed here will act as a comprehensive overview of the roles of cellular senescence in IR injury, which could be of great significance for the design of related studies, or as a guide for potential therapeutic target.
Collapse
|
33
|
Wang L, Dai M, Ge Y, Chen J, Wang C, Yao C, Lin Y. EGCG protects the mouse brain against cerebral ischemia/reperfusion injury by suppressing autophagy via the AKT/AMPK/mTOR phosphorylation pathway. Front Pharmacol 2022; 13:921394. [PMID: 36147330 PMCID: PMC9489224 DOI: 10.3389/fphar.2022.921394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke remains one of the leading reasons of mortality and physical disability worldwide. The treatment of cerebral ischemic stroke faces challenges, partly due to a lack of effective treatments. In this study, we demonstrated that autophagy was stimulated by transient middle cerebral artery occlusion/reperfusion (MCAO/R) and oxygen-glucose deprivation/reoxygenation (OGD/R). Treatment with (−)-epigallocatechin-3-gallate (EGCG), a bioactive ingredient in green tea, was able to mitigate cerebral ischemia/reperfusion injury (CIRI), given the evidence that EGCG administration could reduce the infarct volume and protect poststroke neuronal loss in MCAO/R mice in vivo and attenuate cell loss in OGD/R-challenged HT22 cells in vitro through suppressing autophagy activity. Mechanistically, EGCG inhibited autophagy via modulating the AKT/AMPK/mTOR phosphorylation pathway both in vivo and in vitro models of stroke, which was further confirmed by the results that the administration of GSK690693, an AKT/AMPK inhibitor, and rapamycin, an inhibitor of mTOR, reversed aforementioned changes in autophagy and AKT/AMPK/mTOR signaling pathway. Overall, the application of EGCG relieved CIRI by suppressing autophagy via the AKT/AMPK/mTOR phosphorylation pathway.
Collapse
Affiliation(s)
- Li Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Maosha Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangyang Ge
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiayi Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengye Yao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Chengye Yao, ; Yun Lin,
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Chengye Yao, ; Yun Lin,
| |
Collapse
|
34
|
Liu Y, Yang G, Cui W, Zhang Y, Liang X. Regulatory mechanisms of tetramethylpyrazine on central nervous system diseases: A review. Front Pharmacol 2022; 13:948600. [PMID: 36133805 PMCID: PMC9483103 DOI: 10.3389/fphar.2022.948600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Central nervous system (CNS) diseases can lead to motor, sensory, speech, cognitive dysfunction, and sometimes even death. These diseases are recognized to cause a substantial socio-economic impact on a global scale. Tetramethylpyrazine (TMP) is one of the main active ingredients extracted from the Chinese herbal medicine Ligusticum striatum DC. (Chuan Xiong). Many in vivo and in vitro studies have demonstrated that TMP has a certain role in the treatment of CNS diseases through inhibiting calcium ion overload and glutamate excitotoxicity, anti-oxidative/nitrification stress, mitigating inflammatory response, anti-apoptosis, protecting the integrity of the blood-brain barrier (BBB) and facilitating synaptic plasticity. In this review, we summarize the roles and mechanisms of action of TMP on ischemic cerebrovascular disease, spinal cord injury, Parkinson’s disease, Alzheimer’s disease, cognitive impairments, migraine, and depression. Our review will provide new insights into the clinical applications of TMP and the development of novel therapeutics.
Collapse
Affiliation(s)
- Yue Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guang Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenqiang Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunling Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yunling Zhang, ; Xiao Liang,
| | - Xiao Liang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yunling Zhang, ; Xiao Liang,
| |
Collapse
|
35
|
Neuroprotective Effect and Possible Mechanisms of Ginsenoside-Rd for Cerebral Ischemia/Reperfusion Damage in Experimental Animal: A Meta-Analysis and Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7650438. [PMID: 36092162 PMCID: PMC9458376 DOI: 10.1155/2022/7650438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 11/18/2022]
Abstract
Ischemic stroke, the most common type of stroke, can lead to a long-term disability with the limitation of effective therapeutic approaches. Ginsenoside-Rd (G-Rd) has been found as a neuroprotective agent. In order to investigate and discuss the neuroprotective function and underlying mechanism of G-Rd in experimental animal models following cerebral ischemic/reperfusion (I/R) injury, PubMed, Embase, SinoMed, and China National Knowledge Infrastructure were searched from their inception dates to May 2022, with no language restriction. Studies that G-Rd was used to treat cerebral I/R damage in vivo were selected. A total of 18 articles were included in this paper, and it was showed that after cerebral I/R damage, G-Rd administration could significantly attenuate infarct volume (19 studies, SMD = −1.75 [−2.21 to − 1.30], P < 0.00001). Subgroup analysis concluded that G-Rd at the moderate doses of >10- <50 mg/kg reduced the infarct volume to the greatest extent, and increasing the dose beyond 50 mg/kg did not produce better results. The neuroprotective effect of G-Rd was not affected by other factors, such as the animal species, the order of administration, and the ischemia time. In comparison with the control group, G-Rd administration could improve neurological recovery (lower score means better recovery: 14 studies, SMD = −1.50 [−2.00 to − 1.00], P < 0.00001; higher score means better recovery: 8 studies, SMD = 1.57 [0.93 to 2.21], P < 0.00001). In addition, this review suggested that G-Rd in vivo can antagonize the reduced oxidative stress, regulate Ca2+, and inhibit inflammatory, resistance to apoptosis, and antipyroptosis on cerebral I/R damage. Collectively, G-Rd is a promising natural neuroprotective agent on cerebral I/R injury with unique advantages and a clear mechanism of action. More clinical randomized, blind-controlled trials are also needed to confirm the neuroprotective effect of G-Rd on cerebral I/R injury.
Collapse
|
36
|
Baixauli-Martín J, Aliena-Valero A, Castelló-Ruiz M, Burguete MC, López-Morales MA, Muñoz-Espín D, Torregrosa G, Salom JB. Brain Cell Senescence: A New Therapeutic Target for the Acute Treatment of Ischemic Stroke. J Neuropathol Exp Neurol 2022; 81:614-620. [PMID: 35763058 DOI: 10.1093/jnen/nlac048] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Aging is a major risk factor for cerebral infarction. Since cellular senescence is intrinsic to aging, we postulated that stroke-induced cellular senescence might contribute to neural dysfunction. Adult male Wistar rats underwent 60-minute middle cerebral artery occlusion and were grouped according to 3 reperfusion times: 24 hours, 3, and 7 days. The major biomarkers of senescence: 1) accumulation of the lysosomal pigment, lipofuscin; 2) expression of the cell cycle arrest markers p21, p53, and p16INK4a; and 3) expression of the senescence-associated secretory phenotype cytokines interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), and interleukin-1β (IL-1β) were investigated in brain samples. Lipofuscin accumulation was scarce at the initial stage of brain damage (24 hours), but progressively increased until it reached massive distribution at 7 days post-ischemia. Lipofuscin granules (aggresomes) were mainly confined to the infarcted areas, that is parietal cortex and adjacent caudate-putamen, which were equally affected. The expression of p21, p53, and p16INK4a, and that of IL-6, TNF-α, and IL-1β, was significantly higher in the ischemic hemisphere than in the non-ischemic hemisphere. These data indicate that brain cell senescence develops during acute ischemic infarction and suggest that the acute treatment of ischemic stroke might be enhanced using senolytic drugs.
Collapse
Affiliation(s)
- Júlia Baixauli-Martín
- From the Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Valencia, Valencia, Spain
| | - Alicia Aliena-Valero
- From the Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - María Castelló-Ruiz
- From the Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Facultad de Ciencias Biológicas, Universidad de Valencia, Valencia, Spain
| | - María C Burguete
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Valencia, Valencia, Spain
| | - Mikahela A López-Morales
- From the Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Daniel Muñoz-Espín
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK
| | - Germán Torregrosa
- From the Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Juan B Salom
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Valencia, Valencia, Spain
- From the Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| |
Collapse
|
37
|
Roesel MJ, Sharma NS, Schroeter A, Matsunaga T, Xiao Y, Zhou H, Tullius SG. Primary Graft Dysfunction: The Role of Aging in Lung Ischemia-Reperfusion Injury. Front Immunol 2022; 13:891564. [PMID: 35686120 PMCID: PMC9170999 DOI: 10.3389/fimmu.2022.891564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/21/2022] [Indexed: 01/14/2023] Open
Abstract
Transplant centers around the world have been using extended criteria donors to remedy the ongoing demand for lung transplantation. With a rapidly aging population, older donors are increasingly considered. Donor age, at the same time has been linked to higher rates of lung ischemia reperfusion injury (IRI). This process of acute, sterile inflammation occurring upon reperfusion is a key driver of primary graft dysfunction (PGD) leading to inferior short- and long-term survival. Understanding and improving the condition of older lungs is thus critical to optimize outcomes. Notably, ex vivo lung perfusion (EVLP) seems to have the potential of reconditioning ischemic lungs through ex-vivo perfusing and ventilation. Here, we aim to delineate mechanisms driving lung IRI and review both experimental and clinical data on the effects of aging in augmenting the consequences of IRI and PGD in lung transplantation.
Collapse
Affiliation(s)
- Maximilian J Roesel
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Institute of Medical Immunology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Nirmal S Sharma
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Andreas Schroeter
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Regenerative Medicine and Experimental Surgery, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Tomohisa Matsunaga
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Yao Xiao
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Hao Zhou
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
38
|
Kim C, Lee SG, Lim S, Jung M, Kwon SP, Hong J, Kang M, Sohn HS, Go S, Moon S, Lee SJ, Kim JS, Kim BS. A Senolytic-Eluting Coronary Stent for the Prevention of In-Stent Restenosis. ACS Biomater Sci Eng 2022; 8:1921-1929. [PMID: 35416659 DOI: 10.1021/acsbiomaterials.1c01611] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The vast majority of drug-eluting stents (DES) elute either sirolimus or one of its analogues. While limus drugs stymie vascular smooth muscle cell (VSMC) proliferation to prevent in-stent restenosis, their antiproliferative nature is indiscriminate and limits healing of the endothelium in stented vessels, increasing the risk of late-stent thrombosis. Oxidative stress, which is associated with vascular injury from stent implantation, can induce VSMCs to undergo senescence, and senescent VSMCs can produce pro-inflammatory cytokines capable of inducing proliferation of neighboring nonsenescent VSMCs. We explored the potential of senolytic therapy, which involves the selective elimination of senescent cells, in the form of a senolytic-eluting stent (SES) for interventional cardiology. Oxidative stress was modeled in vitro by exposing VSMCs to H2O2, and H2O2-mediated senescence was evaluated by cytochemical staining of senescence-associated β-galactosidase activity and qRT-PCR. Quiescent VSMCs were then treated with the conditioned medium (CM) of H2O2-treated VSMCs. Proliferative effects of CM were analyzed by staining for proliferating cell nuclear antigen. Senolytic effects of the first-generation senolytic ABT263 were observed in vitro, and the effects of ABT263 on endothelial cells were also investigated through an in vitro re-endothelialization assay. SESs were prepared by dip coating. Iliofemoral arteries of hypercholesteremic rabbits were implanted with SES, everolimus-eluting stents (EESs), or bare-metal stents (BMSs), and the area of stenosis was measured 4 weeks post-implantation using optical coherence tomography. We found that a portion of H2O2-treated VSMCs underwent senescence, and that CM of H2O2-treated senescent VSMCs triggered the proliferation of quiescent VSMCs. ABT263 reverted H2O2-mediated senescence and the proliferative capacity of senescent VSMC CM. Unlike everolimus, ABT263 did not affect endothelial cell migration and/or proliferation. SES, but not EES, significantly reduced stenosis area in vivo compared with bare-metal stents (BMSs). This study shows the potential of SES as an alternative to current forms of DES.
Collapse
Affiliation(s)
- Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seul-Gee Lee
- Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Songhyun Lim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung-Jun Lee
- Cardiology Division, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jung-Sun Kim
- Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.,Cardiology Division, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea.,Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,Institute of Chemical Processes, Institute of Engineering Research, and BioMAX, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|