1
|
Zhang J, Liu Y, Zhao Y, Zhang S, Xu F, Li F. Synergetic effect of mild hypothermia and antioxidant treatment on ROS-mediated neuron injury under oxygen-glucose deprivation investigated by scanning electrochemical microscopy. Chem Sci 2024; 15:20177-20188. [PMID: 39568945 PMCID: PMC11575619 DOI: 10.1039/d4sc05977h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
Ischemic stroke and reperfusion injury result in neuronal damage and dysfunction associated with oxidative stress, leading to overproduction of cellular reactive oxygen species (ROS) and reactive nitrogen species (RNS). In situ monitoring of the transient ROS and RNS effluxes during rapid pathologic processes is crucial for understanding the relationship between progression of cell damage and role of oxidative stress, and developing the corresponding neuroprotective strategies. Herein, we built oxygen glucose deprivation (OGD) and mild hypothermic (MH) models to mimic the in vitro conditions of ischemic stroke and MH treatment. We used scanning electrochemical microscopy (SECM) to in situ monitor H2O2 and nitric oxide (NO) effluxes from HT22 cells under the OGD and MH treatment conditions. Through quantitative analysis of the H2O2 and NO efflux results, we found that the cellular oxidative stress was primarily manifested through ROS release under OGD conditions, and the MH treatment partially suppressed the excessive H2O2 and NO production induced by reoxygenation. Moreover, the synergistic therapeutic effect of MH with antioxidant treatment significantly reduced the oxidative stress and enhanced the cell survival. Our work reveals the crucial role of oxidative stress in OGD and reperfusion processes, and the effective improvement of cell viability via combination of MH with antioxidants, proposing promising therapeutic interventions for ischemic stroke and reperfusion injury.
Collapse
Affiliation(s)
- Junjie Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University Xi'an 710049 P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University Xi'an 710049 P. R. China
| | - Yulin Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University Xi'an 710049 P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University Xi'an 710049 P. R. China
| | - Yuxiang Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University Xi'an 710049 P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University Xi'an 710049 P. R. China
| | - Siyu Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University Xi'an 710049 P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University Xi'an 710049 P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University Xi'an 710049 P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University Xi'an 710049 P. R. China
| | - Fei Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University Xi'an 710049 P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University Xi'an 710049 P. R. China
| |
Collapse
|
2
|
Miyara SJ, Shinozaki K, Hayashida K, Shoaib M, Choudhary RC, Zafeiropoulos S, Guevara S, Kim J, Molmenti EP, Volpe BT, Becker LB. Differential Mitochondrial Bioenergetics in Neurons and Astrocytes Following Ischemia-Reperfusion Injury and Hypothermia. Biomedicines 2024; 12:1705. [PMID: 39200170 PMCID: PMC11352110 DOI: 10.3390/biomedicines12081705] [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: 03/11/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 09/02/2024] Open
Abstract
The close interaction between neurons and astrocytes has been extensively studied. However, the specific behavior of these cells after ischemia-reperfusion injury and hypothermia remains poorly characterized. A growing body of evidence suggests that mitochondria function and putative transference between neurons and astrocytes may play a fundamental role in adaptive and homeostatic responses after systemic insults such as cardiac arrest, which highlights the importance of a better understanding of how neurons and astrocytes behave individually in these settings. Brain injury is one of the most important challenges in post-cardiac arrest syndrome, and therapeutic hypothermia remains the single, gold standard treatment for neuroprotection after cardiac arrest. In our study, we modeled ischemia-reperfusion injury by using in vitro enhanced oxygen-glucose deprivation and reperfusion (eOGD-R) and subsequent hypothermia (HPT) (31.5 °C) to cell lines of neurons (HT-22) and astrocytes (C8-D1A) with/without hypothermia. Using cell lysis (LDH; lactate dehydrogenase) as a measure of membrane integrity and cell viability, we found that neurons were more susceptible to eOGD-R when compared with astrocytes. However, they benefited significantly from HPT, while the HPT effect after eOGD-R on astrocytes was negligible. Similarly, eOGD-R caused a more significant reduction in adenosine triphosphate (ATP) in neurons than astrocytes, and the ATP-enhancing effects from HPT were more prominent in neurons than astrocytes. In both neurons and astrocytes, measurement of reactive oxygen species (ROS) revealed higher ROS output following eOGD-R, with a non-significant trend of differential reduction observed in neurons. HPT after eOGD-R effectively downregulated ROS in both cells; however, the effect was significantly more effective in neurons. Lipid peroxidation was higher after eOGD-R in neurons, while in astrocytes, the increase was not statistically significant. Interestingly, HPT had similar effects on the reduction in lipoperoxidation after eOGD-R with both types of cells. While glutathione (GSH) levels were downregulated after eOGD-R in both cells, HPT enhanced GSH in astrocytes, but worsened GSH in neurons. In conclusion, neuron and astrocyte cultures respond differently to eOGD-R and eOGD-R + HTP treatments. Neurons showed higher sensitivity to ischemia-reperfusion insults than astrocytes; however, they benefited more from HPT therapy. These data suggest that given the differential effects from HPT in neurons and astrocytes, future therapeutic developments could potentially enhance HPT outcomes by means of neuronal and astrocytic targeted therapies.
Collapse
Affiliation(s)
- Santiago J. Miyara
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Koichiro Shinozaki
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Kei Hayashida
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Muhammad Shoaib
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | | | | | - Sara Guevara
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Junhwan Kim
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Ernesto P. Molmenti
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Surgery, Renown Health, Reno, NV 89502, USA
| | - Bruce T. Volpe
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Lance B. Becker
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| |
Collapse
|
3
|
Xu S, Miao H, Gong L, Feng L, Hou X, Zhou M, Shen H, Chen W. Effects of Different Hypothermia on the Results of Cardiopulmonary Resuscitation in a Cardiac Arrest Rat Model. DISEASE MARKERS 2022; 2022:2005616. [PMID: 35419118 PMCID: PMC9001110 DOI: 10.1155/2022/2005616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/02/2022] [Indexed: 11/17/2022]
Abstract
Objective To investigate the optimal temperature of hypothermia treatment in rats with cardiac arrest caused by ventricular fibrillation (VF) after the return of spontaneous circulation (ROSC). Methods A total of forty-eight male Sprague-Dawley rats were induced by VF through the guidewire with a maximum of 5 mA current and untreated for 8 min. Cardiopulmonary resuscitation (CPR) was performed for 8 min followed by defibrillation (DF). Resuscitated rats were then randomized into the normothermia (37°C) group, milder (35°C) group, mild (33°C) group, or moderate (28°C) group. Hypothermia was immediately induced with surface cooling. The target temperature was maintained for 4 h before rewarming to 37 ± 0.5°C. Moreover, at the end of the 4 h, a rat in each group was randomly selected to be sacrificed for the cerebral cortex electron microscopy observation (n = 1). The other resuscitated animals were observed for up to 72 h after ROSC (n = 7). Left ventricular ejection fraction (LVEF) and left ventricular end diastolic volume (LVEDV) were measured. Survival time, survival rate, and neurological deficit score (NDS) were recorded for 72 h. Results During hypothermia, higher LVEF was observed in the hypothermia groups when compared with normothermia group (35°C vs. 37°C, p < 0.05, 33°C and 28°C vs. 37°C, p < 0.01). Among the hypothermia groups, LVEF was higher in the 28°C group than that of 35°C (p < 0.05). However, both the heart rate (HR) (p < 0.01) and LVEDV (28°C vs. 35°C, p < 0.01, 28°C vs. 37°C and 33°C, p < 0.05) were lowest in the 28°C group when compared with the other groups. There were no significant differences of LVEF and LVEDV between the group 35°C and 33°C (p > 0.05). After rewarming, the LVEF of 35°C group was higher than that of group 37°C, 33°C, and 28°C (35°C vs. 37°C and 28°C, p < 0.01, 35°C vs. 33°C, p < 0.05). Group 35°C and 33°C resulted in longer survival (p < 0.01), higher survival rate (p < 0.01), and lower NDS (35°C vs. 37°C and 28°C, p < 0.01, 33°C vs. 37°C and 28°C, p < 0.05) compared with the group 37°C and 28°C. The extent of damage to cerebral cortex cells in group of 35°C and 33°C was lighter than that in group of 37°C and 28°C. The 35°C group spent less time in the process of cooling and rewarming than the group 33°C and 28°C (p < 0.01). Conclusions An almost equal protective effect of milder hypothermia (35°C) and mild hypothermia (33°C) in cardiac arrest (CA) rats was achieved with more predominant effect than moderate hypothermia (28°C) and normothermia (37°C). More importantly, shorter time spent in cooling and rewarming was required in the 35°C group, indicating its potential clinical application. These findings support the possible use of milder hypothermia (35°C) as a therapeutic agent for postresuscitation.
Collapse
Affiliation(s)
- Shaohua Xu
- Nankai University School of Medicine, Tianjin, China
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hui Miao
- The 3rd Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Liming Gong
- Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Lijie Feng
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xuliang Hou
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Manhong Zhou
- Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Hong Shen
- Nankai University School of Medicine, Tianjin, China
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wei Chen
- The 1st Medical Center of Chinese PLA General Hospital, Beijing, China
- The 3rd Medical Center of Chinese PLA General Hospital, Beijing, China
- Hainan Hospital of Chinese PLA General Hospital, Hainan, China
| |
Collapse
|
4
|
Cao M, Zhao Y, He H, Yue R, Pan L, Hu H, Ren Y, Qin Q, Yi X, Yin T, Ma L, Zhang D, Huang X. New Applications of HBOC-201: A 25-Year Review of the Literature. Front Med (Lausanne) 2021; 8:794561. [PMID: 34957164 PMCID: PMC8692657 DOI: 10.3389/fmed.2021.794561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/05/2021] [Indexed: 01/10/2023] Open
Abstract
If not cured promptly, tissue ischemia and hypoxia can cause serious consequences or even threaten the life of the patient. Hemoglobin-based oxygen carrier-201 (HBOC-201), bovine hemoglobin polymerized by glutaraldehyde and stored in a modified Ringer's lactic acid solution, has been investigated as a blood substitute for clinical use. HBOC-201 was approved in South Africa in 2001 to treat patients with low hemoglobin (Hb) levels when red blood cells (RBCs) are contraindicated, rejected, or unavailable. By promoting oxygen diffusion and convective oxygen delivery, HBOC-201 may act as a direct oxygen donor and increase oxygen transfer between RBCs and between RBCs and tissues. Therefore, HBOC-201 is gradually finding applications in treating various ischemic and hypoxic diseases including traumatic hemorrhagic shock, hemolysis, myocardial infarction, cardiopulmonary bypass, perioperative period, organ transplantation, etc. However, side effects such as vasoconstriction and elevated methemoglobin caused by HBOC-201 are major concerns in clinical applications because Hbs are not encapsulated by cell membranes. This study summarizes preclinical and clinical studies of HBOC-201 applied in various clinical scenarios, outlines the relevant mechanisms, highlights potential side effects and solutions, and discusses the application prospects. Randomized trials with large samples need to be further studied to better validate the efficacy, safety, and tolerability of HBOC-201 to the extent where patient-specific treatment strategies would be developed for various clinical scenarios to improve clinical outcomes.
Collapse
Affiliation(s)
- Min Cao
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yong Zhao
- Anesthesiology, Southwest Medicine University, Luzhou, China
| | - Hongli He
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruiming Yue
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lingai Pan
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Huan Hu
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yingjie Ren
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qin Qin
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xueliang Yi
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Yin
- Surgical Department, Chengdu Second People's Hospital, Chengdu, China
| | - Lina Ma
- Health Inspection and Quarantine, Chengdu Medical College, Chengdu, China
| | - Dingding Zhang
- Sichuan Provincial Key Laboratory for Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaobo Huang
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
5
|
Yao S, Wang L, Chen Q, Lu T, Pu X, Luo C. The effect of mild hypothermia plus rutin on the treatment of spinal cord injury and inflammatory factors by repressing TGF-β/smad pathway. Acta Cir Bras 2021; 36:e360307. [PMID: 33978063 PMCID: PMC8112105 DOI: 10.1590/acb360307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/20/2021] [Indexed: 02/07/2023] Open
Abstract
Purpose To probe the mechanism of mild hypothermia combined with rutin in the
treatment of spinal cord injury (SCI). Methods Thirty rats were randomized into the following groups: control, sham, model,
mild hypothermia (MH), and mild hypothermia plus rutin (MH+Rutin). We used
modified Allen’s method to injure the spinal cord (T10) in rats, and then
treated it with MH or/and rutin immediately. BBB scores were performed on
all rats. We used HE staining for observing the injured spinal cord tissue;
ELISA for assaying TNF-α, IL-1β, IL-8, Myeloperoxidase (MPO), and
Malondialdehyde (MDA) contents; Dihydroethidium (DHE) for measuring the
reactive oxygen species (ROS) content; flow cytometry for detecting
apoptosis; and both RT-qPCR and Western blot for determining the expression
levels of TGF-β/Smad pathway related proteins (TGF-β, Smad2, and Smad3). Results In comparison with model group, the BBB score of MH increased to a certain
extent and MH+Rutin group increased more than MH group (p < 0.05). After
treatment with MH and MH+Rutin, the inflammatory infiltration diminished. MH
and MH+Rutin tellingly dwindled TNF-β, MDA and ROS contents (p < 0.01),
and minified spinal cord cell apoptosis. MH and MH+Rutin could patently
diminished TGF-β1, Smad2, and Smad3 expression (p < 0.01). Conclusions MH+Rutin can suppress the activation of TGF-β/Smad pathway, hence repressing
the cellular inflammatory response after SCI.
Collapse
|
6
|
Liu X, Rao S, Wang J. Intravenous thrombolysis in combination with mild hypothermia therapy in the treatment of acute cerebral infarction. Pak J Med Sci 2019; 35:1161-1166. [PMID: 31372161 PMCID: PMC6659097 DOI: 10.12669/pjms.35.4.311] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective To investigate the efficacy of recombinant tissue plasminogen activator (rt-PA) intravenous thrombolysis in combination with mild hypothermia therapy in the treatment of acute cerebral infarction. Methods One hundred and thirty-two patients with acute cerebral infarction who were admitted to our hospital were selected and grouped into a control group and an observation group, 66 each group. Patients in the control group were given conventional treatment in combination with local mild hypothermia therapy, and patients in the observation group were given rt-PA intravenous thrombolysis on the basis of conventional treatment and local mild hypothermia therapy. National institute of health stroke scale (NIHSS) score and intracranial pressure (ICP) of the two groups before and after treatment was recorded. The efficacy of the two groups was evaluated. The modified Rankin scale (MRS) score was followed up for three months. The blood samples of the patients were collected before and after thrombolysis. Superoxide dismutase (SOD) and malondialdehyde (MDA) levels in the plasma were detected. Results The NIHSS score of the two groups decreased in the 1st, 3rd and 7th day after treatment compared to before treatment (p<0.05), but the NIHSS score of the two groups had no significant difference at different time points after treatment (p>0.05). The ICP of the two groups decreased in the 1st, 3rd and 7th day after treatment compared to before treatment (p<0.05), and the decrease of ICP of the observation group was more significant than that of the control group at the same time point (1st, 3rd and 7th day after treatment) (p<0.05). The clinical efficacy of the observation group was higher than that of the control group after treatment, and the difference was statistically significant (p<0.05). The MDA concentration of both groups decreased at different time points after treatment (p<0.05), but the SOD concentration increased (p<0.05). The MDA concentration of the observation group was lower than that of the control group at different time points after treatment (p<0.05), and the SOD concentration of the observation group was higher than that of the control group (p<0.05). Conclusion rt-PA intravenous thrombolysis in combination with mild hypothermia therapy has significant efficacy in the treatment of acute cerebral infarction. It can effectively relieve neurological function. Its action mechanism may be realized by relieving oxidative stress response.
Collapse
Affiliation(s)
- Xiaoying Liu
- Xiaoying Liu Departments of Neurology, Binzhou People's Hospital, Shandong, 256610, China
| | - Shengli Rao
- Shengli Rao Departments of Emergency, Binzhou People's Hospital, Shandong, 256610, China
| | - Jiajia Wang
- Jiajia Wang Departments of Neurology, Binzhou People's Hospital, Shandong, 256610, China
| |
Collapse
|
7
|
Zhu S, Gao X, Huang K, Gu Y, Hu Y, Wu Y, Ji Z, Wang Q, Pan S. Glibenclamide Enhances the Therapeutic Benefits of Early Hypothermia after Severe Stroke in Rats. Aging Dis 2018; 9:685-695. [PMID: 30090656 PMCID: PMC6065285 DOI: 10.14336/ad.2017.0927] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/27/2017] [Indexed: 01/20/2023] Open
Abstract
Glibenclamide (GBC) is an antidiabetic drug that is in a class of medications known as sulfonylureas, which play critical roles in attenuating brain edema and reducing mortality in ischemic stroke patients. Therapeutic hypothermia (TH) is another robust neuroprotectant that prevents brain swelling and improves the neurological outcomes of stroke patients. However, whether the combination of GBC and TH can be used as a reliable neuroprotectant in ischemic stroke remains largely unknown. We used the middle cerebral artery occlusion (MCAO) rat model as well as oxygen and glucose deprivation-reoxygenation (OGD/R) endothelial cells as ischemic stroke models to investigate the efficacy and mechanisms of treating ischemic stroke with the combination of GBC and TH. The serum glucose, mortality rate, neurobehavioral functions, tight junctions, endothelial cells and inflammatory cytokines were evaluated in the stroke models after treatment with GBC, TH or the combination of them. After 5-hour occlusion and subsequent reperfusion, rats exhibited a large volume of hemispheric swelling and a high mortality rate. Stroke rats treated with the combined therapy did not exhibit hypoglycemia. The combination of GBC and TH exhibited synergistic neuroprotective effects in stroke rats that were associated with greater reductions in edema volume, better improvement in neurobehavioral functions, prevention of tight junction loss, and reduction of expression of the inflammatory cytokines COX-2 and iNOS. In OGD/R endothelia cells, the combination reduced endothelial cell death. This study demonstrated that both GBC and TH are neuroprotective after the severe stroke; however, combined therapy with GBC and TH enhanced the efficiency and efficacy of the effects of TH and GBC in the treatment of ischemia. This combined therapy may facilitate the clinical translation of TH management for severe stroke. The combination of GBC and TH seems to be a feasible and promising clinical strategy to alleviate cerebral injury following severe stroke.
Collapse
Affiliation(s)
- Shuzhen Zhu
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,2Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoya Gao
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,2Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kaibin Huang
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yong Gu
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yafang Hu
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongming Wu
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhong Ji
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qing Wang
- 2Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Suyue Pan
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Gao X, Xie H, Zhu S, Yu B, Xian Y, Ouyang Q, Ji Y, Yang X, Wen C, Wang P, Tong Y, Wang Q. The Combination of Human Urinary Kallidinogenase and Mild Hypothermia Protects Adult Rats Against Hypoxic-Ischemic Encephalopathy-Induced Injury by Promoting Angiogenesis and Regeneration. Front Aging Neurosci 2018; 10:196. [PMID: 30050428 PMCID: PMC6050362 DOI: 10.3389/fnagi.2018.00196] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022] Open
Abstract
Objectives: Human Urinary Kallidinogenase (HUK) is a tissue kallikrein that plays neuroprotective role in ischemic conditions via different mechanisms. Mild hypothermia (MH) is another robust neuroprotectant that reduces mortality but does not profoundly ameliorate the neurological outcome in hypoxic-ischemic encephalopathy (HIE) patients. However, whether the combination of HUK and MH can be used as a promising neuroprotective treatment in HIE is unknown. Methods: One-hundred and forty-four adult Wistar rats were randomly divided into five groups: Sham, HIE, HUK, MH and a combination of HUK and MH treatment. The HIE rat model was established by right carotid dissection followed by hypoxia aspiration. The survival curve was created within 7 days, and the neurological severity scores (NSS) were assessed at days 0, 1, 3, and 7. Nissl staining, Terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL), immunofluorescent staining and western blotting were used to evaluate neuronal survival, apoptosis and necrosis, tight-junction proteins Claudin-1 and Zonula occludens-1 (ZO-1), vascular endothelial growth factor (VEGF), doublecortex (DCX), bradykinin receptor B1 (BDKRB1), BDKRB2 and Ki67 staining. Results: The combined treatment rescued all HIE rats from death and had a best survival curve compared to HIE. The Combination also reduced the NSS scores after HIE at days 7, better than HUK or MH alone. The combination of HUK and MH reserved more cells in Nissl staining and inhibited neuronal apoptosis and necrosis as well as significantly attenuated HIE-induced decreases in claudin-1, ZO-1, cyclin D1 and BDKRB1/B2 in comparison to HUK or MH treatment alone. Moreover, the combined treatment increased the expression of VEGF and DCX as well as the number of Ki67-labeled cells. Conclusions: This study demonstrates that both HUK and MH are neuroprotective after HIE insult; however, the combined therapy with HUK and MH enhanced the efficiency and efficacy of either therapy alone in the treatment of HIE, at least partially by promoting angiogenesis and regeneration and rescuing tight-junction loss. The combination of HUK and MH seems to be a feasible and promising clinical strategy to alleviate cerebral injury following HIE insult. Highlights: -The combination of HUK and MH distinctly reduces neurological dysfunction in HIE rats.-HUK enhances the neuroprotective effects of MH in HIE.-MH attenuates tight-junction disruption, upregulates the BDKR B1/2, DCX and cyclin D1.-The combination of MH and HUK enhances the expressions of MH/HUK mediated-BDKR B1/2, DCX, cyclin D1 and Ki67 positive cells.
Collapse
Affiliation(s)
- Xiaoya Gao
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Haiting Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Bin Yu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Xian
- Department of General Intensive Care Unit of Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qian Ouyang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yabin Ji
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaohua Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Chunyan Wen
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Penghua Wang
- Department of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Yufeng Tong
- Structural Genomics Consortium, Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Gao Y, Wang Z, He W, Ma W, Ni X. Mild hypothermia protects neurons against oxygen glucose deprivation via poly (ADP-ribose) signaling. J Matern Fetal Neonatal Med 2017; 32:1633-1639. [PMID: 29278964 DOI: 10.1080/14767058.2017.1413548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Hypothermia is a neuroprotective mechanism that has been validated for use in alleviating neonatal hypoxic-ischemic (HI) brain injury. Nevertheless, it is unclear whether poly (ADP-ribose) (PAR) signaling is involved in hypothermia-induced neuroprotection. In this study, we investigated whether mild hypothermia rescues oxygen glucose deprivation (OGD)-induced cell death by modifying PAR-relative protein expression, such as AIF, PARP-1, and PAR polymer, in primary-cultured hippocampal neurons. METHODS We analyzed neuronal morphology and related protein expression of PAR signaling after OGD followed by mild hypothermia in primary-cultured newborn hippocampal neurons. RESULTS Hypothermic treatment resulted in improved neuronal viability and alleviated DNA damage. Results from the protein assay showed that hypothermia attenuated nuclear translocation of apoptosis-inducing factor (AIF), inhibited overactivation of poly(ADP-ribose) polymerase-1 (PARP-1), and decreased production of PAR polymer induced by PARP-1 activation after OGD. CONCLUSIONS These results showed that mild hypothermia partially protects immature hippocampal neurons against OGD injury in part by interfering with the PAR signaling pathway.
Collapse
Affiliation(s)
- Yubo Gao
- a Department of Anaesthesiology , General Hospital of Ningxia Medical University , Yinchuan , China
| | - Zhihua Wang
- a Department of Anaesthesiology , General Hospital of Ningxia Medical University , Yinchuan , China
| | - Weikun He
- a Department of Anaesthesiology , General Hospital of Ningxia Medical University , Yinchuan , China
| | - Wenjing Ma
- a Department of Anaesthesiology , General Hospital of Ningxia Medical University , Yinchuan , China
| | - Xinli Ni
- a Department of Anaesthesiology , General Hospital of Ningxia Medical University , Yinchuan , China
| |
Collapse
|
10
|
Zhou T, Lin H, Jiang L, Yu T, Zeng C, Liu J, Yang Z. Mild hypothermia protects hippocampal neurons from oxygen-glucose deprivation injury through inhibiting caspase-3 activation. Cryobiology 2017; 80:55-61. [PMID: 29223591 DOI: 10.1016/j.cryobiol.2017.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/18/2017] [Accepted: 12/05/2017] [Indexed: 12/22/2022]
Abstract
Mild hypothermia (MH) is thought to be one of the most effective therapeutic methods to treat hypoxic-ischemic encephalopathy (HIE) after cardiac arrest (CA). However, its precise mechanisms remain unclear. In this research, hippocampal neurons were cultured and treated with mild hypothermia and Ac-DEVD-CHO after oxygen-glucose deprivation (OGD). The activity of caspase-3 was detected, in order to find the precise concentration of Ac-DEVD-CHO with the same protective role in OGD injury as MH treatment. Western blot and immunofluorescence staining were conducted to analyze the effects of MH and Ac-DEVD-CHO on the expressions of caspase-3, caspase-8, and PARP. The neuronal morphology was observed with an optical microscope. The lactic acid dehydrogenase (LDH) release rate, neuronal viability, and apoptotic rate were also detected. We found that MH (32 °C) and Ac-DEVD-CHO (5.96 μMol/L) had equal effects on blocking the activation of caspase-3 and the OGD-induced cleavage of PARP, but neither had any effect on the activation of caspase-8, which goes on to activate caspase-3 in the apoptotic pathway. Meanwhile, both MH and Ac-DEVD-CHO had similar effects in protecting cell morphology, reducing LDH release, and inhibiting OGD-induced apoptosis in neurons. They also similarly improved neuronal viability after OGD. In conclusion, caspase-3 serves as a key intervention point of the key modulation site or regulatory region in MH treatment that protects neuronal apoptosis against OGD injury. Inhibiting the expression of caspase-3 had a protective effect against OGD injury in MH treatment, and caspase-3 activation could be applied to evaluate the neuroprotective effectiveness of MH on HIE.
Collapse
Affiliation(s)
- Tianen Zhou
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Lin
- Hospital of South China Agricultural University, Guangzhou, China
| | - Longyuan Jiang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Yu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chaotao Zeng
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Juanhua Liu
- The Eastern Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zhengfei Yang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Zengcheng District People's Hospital of Guangzhou, Guangzhou, China.
| |
Collapse
|
11
|
Ma SP, Ju F, Zhang YP, Shi X, Zhuang RJ, Xue H, Ma J, Wang L, Cheng BF, Cao H, Feng ZW, Wang M, Yang HJ. Cold-inducible protein RBM3 protects neuroblastoma cells from retinoic acid-induced apoptosis via AMPK, p38 and JNK signaling. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.05.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
12
|
Zhou T, Liang Y, Jiang L, Yu T, Zeng C, Tao E. Mild hypothermia protects against oxygen glucose deprivation/reoxygenation-induced apoptosis via the Wnt/β-catenin signaling pathway in hippocampal neurons. Biochem Biophys Res Commun 2017; 486:1005-1013. [PMID: 28365156 DOI: 10.1016/j.bbrc.2017.03.153] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 01/31/2023]
Abstract
Mild hypothermia is thought to be one of the most effective therapies for cerebral ischemia/reperfusion injuries. Our previous research revealed that mild hypothermia inhibits the activation of caspase-3 and protects against oxygen glucose deprivation/reoxygenation (OGD/R)-induced injury in hippocampal neurons. However, the mechanisms behind the activation of caspase-3 remain unclear. The aims of this study were to determine whether the protective effects of mild hypothermia were exerted through the Wnt/β-catenin signaling pathway. We found that, under OGD/R conditions, the pathway was down regulated, but mild hypothermia induced the reactivation of the Wnt/β-catenin signaling pathway, which had been suppressed by OGD/R injury. Mild hypothermia also caused the down regulation of the expression of apoptosis promoting proteins (Bax cleaved caspase-3), up-regulated the expression of apoptosis inhibiting proteins (Bcl-2), and ameliorated OGD/R injury-induced apoptosis. The protective effects of mild hypothermia were blocked by DKK1 (an antagonist of the canonical Wnt signaling pathway). Taken together, these results indicate that the Wnt/β-catenin signaling pathway mediates the protective effects of mild hypothermia against OGD/R-induced apoptosis. Our study provides evidence that mild hypothermia reactivates the Wnt/β-catenin signaling pathway, which is suppressed by OGD/R injury, in hippocampal neurons and protects neurons from OGD/R-induced apoptosis via the reactivation of the Wnt/β-catenin signaling pathway, ultimately suggesting that mild hypothermia could have therapeutic effects on OGD/R-induced apoptosis.
Collapse
Affiliation(s)
- Tianen Zhou
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yanran Liang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Longyuan Jiang
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Tao Yu
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chaotao Zeng
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Enxiang Tao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
13
|
Singh DP, Barani Lonbani Z, Woodruff MA, Parker TJ, Steck R, Peake JM. Effects of Topical Icing on Inflammation, Angiogenesis, Revascularization, and Myofiber Regeneration in Skeletal Muscle Following Contusion Injury. Front Physiol 2017; 8:93. [PMID: 28326040 PMCID: PMC5339266 DOI: 10.3389/fphys.2017.00093] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/06/2017] [Indexed: 01/01/2023] Open
Abstract
Contusion injuries in skeletal muscle commonly occur in contact sport and vehicular and industrial workplace accidents. Icing has traditionally been used to treat such injuries under the premise that it alleviates pain, reduces tissue metabolism, and modifies vascular responses to decrease swelling. Previous research has examined the effects of icing on inflammation and microcirculatory dynamics following muscle injury. However, whether icing influences angiogenesis, collateral vessel growth, or myofiber regeneration remains unknown. We compared the effects of icing vs. a sham treatment on the presence of neutrophils and macrophages; expression of CD34, von Willebrands factor (vWF), vascular endothelial growth factor (VEGF), and nestin; vessel volume; capillary density; and myofiber regeneration in skeletal after muscle contusion injury in rats. Muscle tissue was collected 1, 3, 7, and 28 d after injury. Compared with uninjured rats, muscles in rats that sustained the contusion injury exhibited major necrosis, inflammation, and increased expression of CD34, vWF, VEGF, and nestin. Compared with the sham treatment, icing attenuated and/or delayed neutrophil and macrophage infiltration; the expression of vWF, VEGF, and nestin; and the change in vessel volume within muscle in the first 7 d after injury (P < 0.05). By contrast, icing did not influence capillary density in muscle 28 d after injury (P = 0.59). The percentage of immature myofibers relative to the total number of fibers was greater in the icing group than in the sham group 28 d after injury (P = 0.026), but myofiber cross-sectional area did not differ between groups after 7 d (P = 0.35) and 28 d (P = 0.30). In conclusion, although icing disrupted inflammation and some aspects of angiogenesis/revascularization, these effects did not result in substantial differences in capillary density or muscle growth.
Collapse
Affiliation(s)
- Daniel P Singh
- Tissue Repair and Regeneration Group, Institute of Health and Biomedical Innovation, Queensland University of Technology Brisbane, QLD, Australia
| | - Zohreh Barani Lonbani
- Tissue Repair and Regeneration Group, Institute of Health and Biomedical Innovation, Queensland University of Technology Brisbane, QLD, Australia
| | - Maria A Woodruff
- Biofabrication and Tissue Morphology Group, Institute of Health and Biomedical Innovation, Queensland University of Technology Brisbane, QLD, Australia
| | - Tony J Parker
- Tissue Repair and Regeneration Group, Institute of Health and Biomedical Innovation, Queensland University of TechnologyBrisbane, QLD, Australia; School of Biomedical Sciences, Queensland University of TechnologyBrisbane, QLD, Australia
| | - Roland Steck
- Medical Engineering Research Facility, Queensland University of Technology Brisbane, QLD, Australia
| | - Jonathan M Peake
- Tissue Repair and Regeneration Group, Institute of Health and Biomedical Innovation, Queensland University of TechnologyBrisbane, QLD, Australia; School of Biomedical Sciences, Queensland University of TechnologyBrisbane, QLD, Australia
| |
Collapse
|