1
|
Abdel-Hameed SS, El-Daly M, Ahmed ASF, Bekhit AA, Heeba GH. Dapoxetine prevents neuronal damage and improves functional outcomes in a model of ischemic stroke through the modulation of inflammation and oxidative stress. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:253-266. [PMID: 37417988 PMCID: PMC10771602 DOI: 10.1007/s00210-023-02601-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023]
Abstract
Stroke is a medical emergency that is associated with substantial mortality and functional disability in adults. The most popular class of antidepressants, selective serotonin reuptake inhibitors SSRIs, have recently been shown in studies to have positive effects on post-stroke motor and cognitive function. Thus, we hypothesized that dapoxetine (DAP), a short-acting SSRI, would be effective against cerebral ischemia/reperfusion injury. Adult male Wister rats (200-250 g) were subjected to a sham operation or bilateral common carotid artery occlusion (BCCAO) for 30 min followed by 24 h of reperfusion to induce global cerebral ischemia/reperfusion (I/R) injury. Rats were treated with vehicle or DAP (30 or 60 mg/kg, i.p.) 1 h before BCCAO. The neurobehavioral performance of rats was assessed. The infarct volume, histopathological changes, oxidative stress parameters, and apoptotic and inflammatory mediators were determined in the brain tissues of euthanized rats. Our results confirmed that DAP significantly ameliorated cerebral I/R-induced neurobehavioral deficits, reduced cerebral infarct volume, and histopathological damage. Moreover, DAP pretreatment reduced lipid peroxidation, caspase-3, and inflammatory mediators (TNF-α and iNOS) compared to I/R-injured rats. Thus, DAP pretreatment potentially improves neurological function, and cerebral damage in cerebral ischemic rats may be partly related to the reduction in the inflammatory response, preservation of oxidative balance, and suppression of cell apoptosis in brain tissues.
Collapse
Affiliation(s)
| | - Mahmoud El-Daly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Amany A Bekhit
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Gehan H Heeba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt.
| |
Collapse
|
2
|
Tonev D, Momchilova A. Oxidative Stress and the Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) Pathway in Multiple Sclerosis: Focus on Certain Exogenous and Endogenous Nrf2 Activators and Therapeutic Plasma Exchange Modulation. Int J Mol Sci 2023; 24:17223. [PMID: 38139050 PMCID: PMC10743556 DOI: 10.3390/ijms242417223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/18/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
The pathogenesis of multiple sclerosis (MS) suggests that, in genetically susceptible subjects, T lymphocytes undergo activation in the peripheral compartment, pass through the BBB, and cause damage in the CNS. They produce pro-inflammatory cytokines; induce cytotoxic activities in microglia and astrocytes with the accumulation of reactive oxygen species, reactive nitrogen species, and other highly reactive radicals; activate B cells and macrophages and stimulate the complement system. Inflammation and neurodegeneration are involved from the very beginning of the disease. They can both be affected by oxidative stress (OS) with different emphases depending on the time course of MS. Thus, OS initiates and supports inflammatory processes in the active phase, while in the chronic phase it supports neurodegenerative processes. A still unresolved issue in overcoming OS-induced lesions in MS is the insufficient endogenous activation of the Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) pathway, which under normal conditions plays an essential role in mitochondria protection, OS, neuroinflammation, and degeneration. Thus, the search for approaches aiming to elevate endogenous Nrf2 activation is capable of protecting the brain against oxidative damage. However, exogenous Nrf2 activators themselves are not without drawbacks, necessitating the search for new non-pharmacological therapeutic approaches to modulate OS. The purpose of the present review is to provide some relevant preclinical and clinical examples, focusing on certain exogenous and endogenous Nrf2 activators and the modulation of therapeutic plasma exchange (TPE). The increased plasma levels of nerve growth factor (NGF) in response to TPE treatment of MS patients suggest their antioxidant potential for endogenous Nrf2 enhancement via NGF/TrkA/PI3K/Akt and NGF/p75NTR/ceramide-PKCζ/CK2 signaling pathways.
Collapse
Affiliation(s)
- Dimitar Tonev
- Department of Anesthesiology and Intensive Care, University Hospital “Tzaritza Yoanna—ISUL”, Medical University of Sofia, 1527 Sofia, Bulgaria
| | - Albena Momchilova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Science, 1113 Sofia, Bulgaria;
| |
Collapse
|
3
|
Li T, Su D, Lu H, Gao Y, Liu Y, Wang S, Hou Y, Qin K, Que X, Chen X, Qin B, Wang Z, Deng Y. Recombinant human brain natriuretic peptide attenuates ischemic brain injury in mice by inhibiting oxidative stress and cell apoptosis via activation of PI3K/AKT/Nrf2/HO-1 pathway. Exp Brain Res 2023; 241:2751-2763. [PMID: 37847304 DOI: 10.1007/s00221-023-06716-4] [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: 06/25/2023] [Accepted: 09/19/2023] [Indexed: 10/18/2023]
Abstract
Ischemic stroke followed by cerebral artery occlusion is a main cause of chronic disability worldwide. Recombinant human brain natriuretic peptide (rhBNP) has been reported to alleviate sepsis-induced cognitive dysfunction and brain I/R injury. However, the function and molecular mechanisms of rhBNP in ischemic brain injury have not been clarified. For establishment of an animal model of ischemic brain injury, C57BL/6 mice were treated with middle cerebral artery occlusion (MCAO) surgery for 1 h and reperfusion for 24 h. After subcutaneous injection of rhBNP into model mice, neurologic deficits were assessed by evaluating behavior of mice according to Longa scoring system, and TTC staining was utilized to determine the brain infarct size of mice. The levels of oxidative stress markers, superoxide dismutase (SOD), catalase (CAT), glutathione (GSH) and malondialdehyde (MDA), were detected in hippocampal tissues of mice by corresponding kits. Cell apoptosis in hippocampus tissues was examined by TUNEL staining. Protein levels of antioxidant enzymes (HO-1 and NQO1) in cerebral cortex, apoptotic markers (Bax, Bcl-2, and cleaved caspase), and PI3K/AKT pathway-associated factors in hippocampus were tested by western blot analysis. The results revealed that injection of rhBNP decreased neurologic deficit scores, the percent of brain water content, and infarct volume. Additionally, rhBNP downregulated MDA level, upregulated the levels of SOD, CAT, and GSH in hippocampus of mice, and increased protein levels of HO-1 and NQO1 in the cortex. Cell apoptosis in hippocampus tissues of model mice was inhibited by rhBNP which was shown as the reduced TUNEL-positive cells, the decreased Bax, cleaved caspase-3, and cleaved caspase-9 protein levels, and the enhanced Bcl-2 protein level. In addition, rhBNP treatment activated the PI3K/AKT signaling pathway and upregulated the protein levels of HO-1 and NRF2. Overall, rhBNP activates the PI3K/AKT/HO-1/NRF2 pathway to attenuate ischemic brain injury in mice after MCAO by suppression of cell apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Tong Li
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - DaJing Su
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - HuaWen Lu
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - YunQing Gao
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - YongGang Liu
- Department of Urology Surgery, Nanning Second People's Hospital, Nanning, 530031, China
| | - ShaoHua Wang
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - YuTing Hou
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - KeMin Qin
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - XianTing Que
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - XiaoPing Chen
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - BaiLing Qin
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - ZiJun Wang
- Department of Neurology, Nanning Second People's Hospital, Nanning, 530031, China
| | - Yan Deng
- Department of Medical Records, Nanning Second People's Hospital, No. 13, Dancun Road, Jiangnan District, Nanning, 530031, Guangxi, China.
| |
Collapse
|
4
|
Schettler VJJ, Schettler E. Beyond cholesterol-pleiotropic effects of lipoprotein apheresis. Ther Apher Dial 2022; 26 Suppl 1:35-40. [PMID: 36468323 DOI: 10.1111/1744-9987.13857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 03/21/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is a leading cause of mortality worldwide, which is caused mainly by atherosclerosis, a chronic inflammatory disease of blood vessels. Therefore, atherosclerosis represents a complex disorder, which induces damage or imbalance on different levels: for example, genes, cytokines, lipoproteins, cells, vessels, and organs. Lipoprotein apheresis (LA) is a well-established extracorporeal treatment of severe hyperlipoproteinemia. In addition, LA may have simultaneously crucial effects on many other atherogenic factors during the treatments, for example, as vascular inflammation, rheology, mobilization of adult stem cells and gene expressions in blood or endothelial cells, which will be discussed in this short review. In addition, stable microRNAs besides tissues also appear in extracellular compartments, for example, vessels, involved in atherosclerotic processes, were found to be reduced by LA treatments. In summary, LA represents a complex therapeutic procedure, that provides an ideal tool for the treatment of complex disorders such as atherosclerosis.
Collapse
Affiliation(s)
| | - Elke Schettler
- BRAVE - Benefit for Research on Arterial Hypertension, Dyslipidemia and Vascular Risk and Education e.V., Göttingen, Germany
| |
Collapse
|
5
|
Zhang X, You LY, Zhang ZY, Jiang DX, Qiu Y, Ruan YP, Mao ZJ. Integrating pharmacological evaluation and computational identification for deciphering the action mechanism of Yunpi-Huoxue-Sanjie formula alleviates diabetic cardiomyopathy. Front Pharmacol 2022; 13:957829. [PMID: 36147338 PMCID: PMC9487204 DOI: 10.3389/fphar.2022.957829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Yunpi-Huoxue-Sanjie (YP-SJ) formula is a Chinese herbal formula with unique advantages for the treatment of diabetic cardiovascular complications, such as Diabetic cardiomyopathy (DCM). However, potential targets and molecular mechanisms remain unclear. Therefore, our research was designed to evaluate rat myocardial morphology, fat metabolism and oxidative stress to verify myocardial protective effect of YP-SJ formula in vivo. And then to explore and validate its probable mechanism through network pharmacology and experiments in vitro and in vivo. Methods: In this study, DCM rats were randomly divided into five groups: control group, model group, and three YP-SJ formula groups (low-dose, middle-dose, and high-dose groups). Experimental rats were treated with 6 g/kg/d, 12 g/kg/d and 24 g/kg/d YP-SJ formula by gavage for 10 weeks, respectively. Cardiac function of rats was measured by high-resolution small-animal imaging system. The cells were divided into control group, high glucose group, high glucose + control serum group, high glucose + dosed serum group, high glucose + NC-siRNA group, high glucose + siRNA-FoxO1 group. The extent of autophagy was measured by flow cytometry, immunofluorescence, and western blotting. Results: It was found that YP-SJ formula could effectively improve cardiac systolic function in DCM rats. We identified 46 major candidate YP-SJ formula targets that are closely related to the progression of DCM. Enrichment analysis revealed key targets of YP-SJ formula related to environmental information processing, organic systems, and the metabolic occurrence of reactive oxygen species. Meanwhile, we verified that YP-SJ formula can increase the expression of forkhead box protein O1 (FoxO1), autophagy-related protein 7 (Atg7), Beclin 1, and light chain 3 (LC3), and decrease the expression of phosphorylated FoxO1 in vitro and in vivo. The results showed that YP-SJ formula could activate the FoxO1 signaling pathway associated with DCM rats. Further experiments showed that YP-SJ formula could improve cardiac function by regulating autophagy. Conclusion: YP-SJ formula treats DCM by modulating targets that play a key role in autophagy, improving myocardial function through a multi-component, multi-level, multi-target, multi-pathway, and multi-mechanism approach.
Collapse
Affiliation(s)
- Xin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
| | - Li-Yan You
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
| | - Ze-Yu Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dong-Xiao Jiang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
| | - Yu Qiu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ye-Ping Ruan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
- *Correspondence: Zhu-Jun Mao, ; Ye-Ping Ruan,
| | - Zhu-Jun Mao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
- *Correspondence: Zhu-Jun Mao, ; Ye-Ping Ruan,
| |
Collapse
|
6
|
Zhang J, Gao Y, Zhang L, Zhang C, Zhao Y, Zhang Y, Li S, Chang C, Zhang X, Yang G. Alpha-Lipoic Acid Attenuates MPTP/MPP +-Induced Neurotoxicity: Roles of SIRT1-Dependent PGC-1α Signaling Pathways. Neurotox Res 2022; 40:410-419. [PMID: 35146598 DOI: 10.1007/s12640-022-00479-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/31/2022]
Abstract
Accumulated oxidative damage plays key roles in the pathogenesis of Parkinson's disease (PD). Silent mating type information regulation 2 homolog 1 (SIRT1), a class III histone deacetylase, can directly activate peroxisome proliferator-activated receptor-c coactivator-1α (PGC-1α) and attenuate oxidative stress. Alpha-lipoic acid (ALA) is a natural antioxidant that has been demonstrated to protect PC12 cells against 1-methyl-4-phenylpyridinium (MPP+). However, the underlying mechanisms related to changes in cell signaling cascades are not fully understood. In the present study, the neuroprotective effect of ALA and the potential role of ALA in the SIRT1 pathway was investigated in vitro and in a mouse model of PD. A Cell Counting Kit-8 (CCK-8) assay was performed to detect the SY5Y-SH cell viability. Immunohistochemistry, quantitative real-time polymerase chain reaction and western blot assays were used to evaluate the expression of tyrosine hydroxylase (TH), SIRT1, and PGC-1α in vivo and in vitro. Intracellular reactive oxygen species (ROS) production and tissue SOD and MDA were detected by the corresponding assay kits. The results showed that ALA notably prevented oxidative stress and neurotoxicity in vivo and in vitro against 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)/MPP+. Furthermore, ALA significantly increased the expression of SIRT1 and PGC-1α in vivo and in vitro in MPTP/MPP+-induced models, which was reversed by the SIRT1 inhibitor EX527. These results suggested that ALA prevented oxidative stress and that neurotoxicity was involved in the upregulation of SIRT1 and PGC-1α in PD mice.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 05000, People's Republic of China
| | - Ya Gao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 05000, People's Republic of China
| | - Lan Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Cong Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Yuan Zhao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 05000, People's Republic of China
| | - Yidan Zhang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 05000, People's Republic of China
| | - Shuyue Li
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 05000, People's Republic of China
| | - Cui Chang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 05000, People's Republic of China
| | - Xiangjian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China. .,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China.
| | - Guofeng Yang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 05000, People's Republic of China.
| |
Collapse
|
7
|
Özevren H, Deveci E, Tuncer MC. The effect of rosmarinic acid on deformities occurring in brain tissue by craniectomy method. Histopathological evaluation of IBA-1 and GFAP expressions. Acta Cir Bras 2020; 35:e202000406. [PMID: 32578724 PMCID: PMC7307720 DOI: 10.1590/s0102-865020200040000006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/09/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To investigate the role of Rosmarinic acid (RA) in the prevention of traumatic brain injury and the immunohistochemical analysis of IBA-1 and GFAP expressions. METHODS Healthy male rats were randomly divided into 3 groups consisting of 10 rats. Groups were as follows; control group, traumatic brain injury (TBI) group, and TBI+RA group. After traumatic brain injury, blood samples were taken from the animals and analyzed with various biochemical markers. And then IBA-1 and GFAP expressions were evaluated immunohistochemically. RESULTS Significant results were obtained in all biochemical parameters between groups. Immunohistochemical sections showed IBA-1 not only in microglia and macrophage activity but also in degenerative neurons in blood vessel endothelial cells. However, GFAP reaction and post-traumatic rosmarinic acid administration showed positive expression in astrocytes with regular structure around the blood vessel. CONCLUSION Rosmarinic acid in blood vessel endothelial cells showed that preserving the integrity of astrocytic structure in the blood brain barrier may be an important antioxidant.
Collapse
Affiliation(s)
- Hüseyin Özevren
- Associate Professor, Department of Neurosurgery , Faculty of
Medicine , Dicle University , Diyarbakır , Turkey . Technical procedures, manuscript
preparation and writing, final approval
| | - Engin Deveci
- PhD, Professor, Department of Histology and Embryology , Faculty
of Medicine , Dicle University , Diyarbakır , Turkey . Technical procedures,
histopathological examinations, manuscript preparation and writing, final
approval
| | - Mehmet Cudi Tuncer
- PhD, Professor, Department of Anatomy , Faculty of Medicine ,
Dicle University , Diyarbakır , Turkey . Technical procedures, histopathological
examinations, manuscript preparation and writing, final approval
| |
Collapse
|
8
|
Liang S, Xu Z, Ruan Y, Niu T, Guo W, Jiang W, Hou J. Isoquercitrin Attenuates Renal Ischemia/Reperfusion Injury Through Antioxidation, Anti-inflammation, and Antiapoptosis in Mice. Transplant Proc 2020; 52:1014-1019. [PMID: 32115238 DOI: 10.1016/j.transproceed.2019.12.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 11/09/2019] [Accepted: 12/15/2019] [Indexed: 01/29/2023]
Abstract
Renal ischemia-reperfusion injury (RIRI) occurs after several surgical procedures such as kidney transplantation and partial nephrectomy. Isoquercitrin (IQ) exhibited protective effects in cerebral ischemia-reperfusion injury. In the present study, we aimed to evaluate the effects of IQ on the prevention of RIRI. The mouse model of RIRI was induced by 30-minute clamping of the left renal pedicle after excising of the right kidney, followed by 24-hour reperfusion. Thirty mice were randomly divided into the following 3 groups: sham operation, RIRI model group, and IQ pretreatment + RIRI. Serum creatinine and blood urea nitrogen (BUN) were used for evaluating renal function. Kidney cell apoptosis was measured by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining. Moreover, the pro-inflammatory cytokines (TNF-α, IL-6), the oxidative stress associated factors (malondialdehyde, superoxide dismutase), and the apoptotic factors (Bcl-2, Bax) were assessed. After RIRI, BUN, creatinine, TNF-α, IL-6, malondialdehyde, and Bax were significantly increased, and levels of superoxide dismutase and Bcl-2/Bax ratio and Bcl-2 expression were decreased markedly. As expect, IQ reversed these changes. These data indicate that IQ plays a protective role during RIRI, which may be partially mediated through the actions of antioxidation, anti-inflammation, and antiapoptosis.
Collapse
Affiliation(s)
- Sudong Liang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhen Xu
- Department of Urology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Yashi Ruan
- Department of Urology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Tianli Niu
- Department of Urology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Wei Guo
- Department of Urology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Wei Jiang
- Department of Urology, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
9
|
Shi Y, Qiu X, Dai M, Zhang X, Jin G. Hyperoside Attenuates Hepatic Ischemia-Reperfusion Injury by Suppressing Oxidative Stress and Inhibiting Apoptosis in Rats. Transplant Proc 2019; 51:2051-2059. [PMID: 31399183 DOI: 10.1016/j.transproceed.2019.04.066] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/22/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Hepatic ischemia-reperfusion (IR) injury is a serious complication of many clinical conditions, which may lead to liver or multiple organ failure. Hyperoside, a flavonoid compound, has been reported to protect against myocardial and cerebral injury induced by IR. This study aimed to investigate the protective effects of hyperoside on hepatic IR injury in rats. METHODS Using the 70% hepatic IR injury model, we divided 32 male Wistar rats into 4 groups (n = 8): sham-operated, IR+saline (saline/p.o.), IR+vehicle (carboxy methyl cellulose/p.o.), and IR+hyperoside (50 mg/kg/d/p.o.). At 24 hours after reperfusion, blood and liver tissue were collected. The effects of hyperoside on hepatic IR injury were assessed through tests of serum transaminase, hepatic histopathology, and measurement of markers of oxidative stress and apoptosis. RESULTS Pretreatment with hyperoside protected the liver from IR injury by a reduction in serum aspartate aminotransferase/alanine aminotransferase levels and a decrease in the severity of histologic changes. Hyperoside treatment also decreased the activity of malondialdehyde, increased the activities of superoxide dismutase and glutathione peroxidase, up-regulated the expression of heme oxygenase 1 and NAD(P)H:quinone oxidoreductase 1, and reduced the apoptotic index after IR injury. A decrease in the expression of caspase-3 and an increase in the ratio of B cell lymphoma 2 to B cell lymphoma 2-associated X also were observed. CONCLUSION Hyperoside has a protective effect on hepatic IR injury in rats, which may be due to its antioxidant and antiapoptotic properties.
Collapse
Affiliation(s)
- Yaoping Shi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaoxia Qiu
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Mengjun Dai
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xuebin Zhang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Guangxin Jin
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
10
|
Zhao Q, Wang X, Chen A, Cheng X, Zhang G, Sun J, Zhao Y, Huang Y, Zhu Y. Rhein protects against cerebral ischemic‑/reperfusion‑induced oxidative stress and apoptosis in rats. Int J Mol Med 2018; 41:2802-2812. [PMID: 29436613 PMCID: PMC5846655 DOI: 10.3892/ijmm.2018.3488] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 02/08/2018] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the protective effects of rhein on cerebral ischemic/reperfusion (I/R) injury in rats. The present study focused on the effect of rhein on oxidative stress and apoptotic factors, which are considered to serve an important role in the onset of I/R injury. Sprague-Dawley rats were subjected to middle cerebral artery occlusion. Neurological functional scores (NFSs) were evaluated according to the Zea Longa's score criteria and the area of brain infarct was determined by triphenyltetrazolium chloride staining. The morphology of the nerve cells in the cortex was observed following hematoxylin and eosin staining. In addition, levels of oxidative stress were assessed by measuring the levels of superoxide dismutase (SOD), glutathione-peroxidase (GSH-Px), catalase (CAT) and malondialdehyde (MDA). Levels of B-cell lymphoma-2 (Bcl-2), apoptosis regulator Bax (BAX), caspase-9, caspase-3 and cleaved caspase-3 expression were analyzed using western blot analysis. Levels of caspase-9 and caspase-3 mRNA expression were obtained using reverse transcription-quantitative polymerase chain reaction. The results revealed that treatment with 50 or 100 mg/kg rhein significantly improved the NFS and markedly attenuated the area of infarction. Rhein also significantly reduced the content of MDA and significantly increased SOD, GSH-Px and CAT activity. Western blot analysis indicated that rhein significantly decreased the expression of BAX and enhanced the expression of Bcl-2. Compared with the I/R group, levels of caspase-9, caspase-3 and cleaved caspase-3 protein expression were significantly decreased in the rhein treatment groups. Additionally, rhein treatment significantly reduced levels of caspase-9 and caspase-3 mRNA expression. These results suggest that rhein exhibits protective effects during cerebral I/R injury and its underlying mechanism of action may involve the inhibition of oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Qipeng Zhao
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiaobo Wang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Ailing Chen
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiuli Cheng
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Guoxin Zhang
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jianmin Sun
- College of Basic Medicine, Yinchuan, Ningxia 750004, P.R. China
| | - Yunsheng Zhao
- Ningxia Hui Modern Medicine Engineering Research Center, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu Huang
- Ningxia Hui Modern Medicine Engineering Research Center, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yafei Zhu
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
11
|
Cui HY, Zhang XJ, Yang Y, Zhang C, Zhu CH, Miao JY, Chen R. Rosmarinic acid elicits neuroprotection in ischemic stroke via Nrf2 and heme oxygenase 1 signaling. Neural Regen Res 2018; 13:2119-2128. [PMID: 30323140 PMCID: PMC6199925 DOI: 10.4103/1673-5374.241463] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Rosmarinic acid (RA) can elicit a neuroprotective effect against ischemic stroke, but the precise molecular mechanism remains poorly understood. In this study, an experimental ischemic stroke model was established in CD-1 mice (Beijing Vital River Laboratory Animal Technology, Beijing, China) by occluding the right middle cerebral artery for 1 hour and allowing reperfusion for 24 hours. After intraperitoneally injecting model mice with 10, 20, or 40 mg/kg RA, functional neurological deficits were evaluated using modified Longa scores. Subsequently, cerebral infarct volume was measured using TTC staining and ischemic brain tissue was examined for cell apoptosis with TUNEL staining. Superoxide dismutase activity and malondialdehyde levels were measured by spectrophometry. Expression of heme oxygenase-1 (HO-1), nuclear factor erythroid 2-related factor 2 (Nrf2), Bcl-2, Bax, Akt, and phospho-Ser473 Akt proteins in ischemic brain tissue was detected by western blot, while mRNA levels of Nrf2, HO-1, Bcl-2, and Bax were analyzed using real time quantitative PCR. In addition, HO-1 enzyme activity was measured spectrophotometrically. RA (20 and 40 mg/kg) greatly improved neurological function, reduced infarct volume, decreased cell apoptosis, upregulated Bcl-2 protein and mRNA expression, downregulated Bax protein and mRNA expression, increased HO-1 and Nrf2 protein and mRNA expression, increased superoxide dismutase activity, and decreased malondialdehyde levels in ischemic brain tissue of model mice. However, intraperitoneal injection of a HO-1 inhibitor (10 mg/kg zinc protoporphyrin IX) reversed the neuroprotective effects of RA on HO-1 enzyme activity and Bcl-2 and Bax protein expression. The PI3K/Akt signaling pathway inhibitor LY294002 (10 mM) inhibited Akt phosphorylation, as well as Nrf2 and HO-1 expression. Our findings suggest that RA has anti-oxidative and anti-apoptotic properties that protect against ischemic stroke by a mechanism involving upregulation of Nrf2 and HO-1 expression via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Hai-Ying Cui
- Department of Neurology, Second Hospital of Hebei Medical University; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardiocerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Xiang-Jian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardiocerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Yi Yang
- Department of Neurology, Second Hospital of Hebei Medical University; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardiocerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardiocerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Chun-Hua Zhu
- Department of Neurology, Second Hospital of Hebei Medical University; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardiocerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Jiang-Yong Miao
- Department of Neurology, Second Hospital of Hebei Medical University; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardiocerebrovascular Disease, Shijiazhuang, Hebei Province, China
| | - Rong Chen
- Department of Neurology, Second Hospital of Hebei Medical University; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardiocerebrovascular Disease, Shijiazhuang, Hebei Province, China
| |
Collapse
|
12
|
Xu Z, Zhao K, Han P, Qi X, Zhang W, Niu T. Octreotide Ameliorates Renal Ischemia/Reperfusion Injury via Antioxidation and Anti-inflammation. Transplant Proc 2017; 49:1916-1922. [DOI: 10.1016/j.transproceed.2017.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/01/2017] [Accepted: 05/13/2017] [Indexed: 12/28/2022]
|
13
|
Toward an international consensus—Integrating lipoprotein apheresis and new lipid-lowering drugs. J Clin Lipidol 2017; 11:858-871.e3. [DOI: 10.1016/j.jacl.2017.04.114] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/30/2017] [Accepted: 04/15/2017] [Indexed: 12/11/2022]
|
14
|
Mu F, Duan J, Bian H, Yin Y, Zhu Y, Wei G, Guan Y, Wang Y, Guo C, Wen A, Yang Y, Xi M. Cardioprotective effects and mechanism of Radix Salviae miltiorrhizae and Lignum Dalbergiae odoriferae on rat myocardial ischemia/reperfusion injury. Mol Med Rep 2017; 16:1759-1770. [PMID: 28656200 PMCID: PMC5562082 DOI: 10.3892/mmr.2017.6821] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 04/25/2017] [Indexed: 01/03/2023] Open
Abstract
Radix Salviae miltiorrhizae (SM) and Lignum Dalbergiae odoriferae (DO) are traditional Chinese medicinal herbs used to treat ischemic heart disease and other cardiovascular diseases; however, to the best of our knowledge, there are currently few studies regarding their effects. The present study aimed to investigate the cardioprotective effects of SM and DO during myocardial ischemia/reperfusion (MI/R) injury in rats, and explore the molecular mechanisms that underlie their actions. In the present study, Sprague-Dawley rats were pretreated with SM, the aqueous extract of DO (DOA) and the volatile oil of DO (DOO), either as a monotherapy or in combination for 7 days. Subsequently, the rats were subjected to 30 min of ischemia followed by 180 min of reperfusion. Traditional pharmacodynamic evaluation and metabonomics based on gas chromatography/time-of-flight mass spectrometry were used to identify the therapeutic effects of these traditional Chinese medicines. The results revealed that SM, DOA and DOO monotherapies ameliorated cardiac function, and this effect was strengthened further when used in combined therapies. Among the combined treatments, SM + DOO exhibited the greatest potential (P<0.05) to improve electrocardiogram results and heart rate, reduce the heart weight index and myocardial infarct size, and decrease the levels of creatine kinase-MB and lactate dehydrogenase. In addition, metabonomics-based findings, including the principal component analysis and partial least squares discriminant analysis score plot of the metabolic state in rat serum, provided confirmation for the aforementioned results, verifying that SM + DOO exerted synergistic therapeutic efficacies to exhibit a greater effect on rats with MI/R injury when compared with the other pretreatment groups. Furthermore, the most effective duration of SM + DOO treatment was 30 min and the least effective duration was 180 min. Treatment with SM + DOO also significantly (P<0.01) reduced the number of terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling-positive cells, tumor necrosis factor-α andinterleukin-6 expression, and malondialdehyde content, and increased the serum and tissue activity of superoxide dismutase. These results indicated that the combined effects of SM + DOO may be more effective compared with the single pretreatments against MI/R injury in rats. This effect may be achieved partly through anti-apoptotic, antioxidant and anti-inflammatory activities. Therefore, SM + DOO may be considered an effective and promising novel strategy for the prophylaxis and treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Fei Mu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jialin Duan
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Haixu Bian
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ying Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yanrong Zhu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Guo Wei
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yue Guan
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yanhua Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yong Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Miaomiao Xi
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
15
|
Li XJ, Liang L, Shi HX, Sun XP, Wang J, Zhang LS. Neuroprotective effects of curdione against focal cerebral ischemia reperfusion injury in rats. Neuropsychiatr Dis Treat 2017; 13:1733-1740. [PMID: 28721054 PMCID: PMC5501624 DOI: 10.2147/ndt.s139362] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Curdione is one of the most highly concentrated component of the active constituents in E-zhu, which has been reported to possess a variety of activities. However, the pharmacologic neuroprotective activity of curdione has not been evaluated. The present study aimed to investigate the protective effect of curdione on focal cerebral ischemia reperfusion-induced injury in rats and further exploring the underlying mechanisms. MATERIALS AND METHODS Adult male Sprague Dawley rats were subjected to middle cerebral artery occlusion (MCAO) surgery for 2 h, followed by reperfusion stage. All animals received treatment once a day for 7 days before surgery and 14 days from 4 h after the reperfusion started. The neurological deficit test and Morris water maze test were performed at 1, 4, 7 and 14 days after MCAO. The infarct size of animals was determined by the 2,3,5-triphenyltetrazolium chloride staining, and pathological brain damage was estimated by hematoxylin-eosin staining. The malonaldehyde (MDA) levels and the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-PX) were detected by enzyme-linked immunosorbent assay. Expression of apoptotic proteins was measured by Western blot. RESULTS Our results showed that curdione could significantly reduce the infarct size and neurological deficits, promote cognitive function recovery and recover neuronal morphologic damages in MCAO rats. It also blocked the increase of MDA content and elevated the activities of SOD, CAT and GSH-PX. Moreover, curdione attenuated the expression of Cyt-C, c-caspase-3 and c-caspase-9 increased the Bcl-2/Bax ratio and hence decreased the cellular apoptosis. CONCLUSION Curdione possessed potential neuroprotective effect on rats in the MCAO model. The anti-oxidative and anti-apoptotic properties may be involved in the underlying mechanisms.
Collapse
Affiliation(s)
- Xing-Jie Li
- Health Management Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Li Liang
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Hong-Xia Shi
- Health Management Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Xiao-Ping Sun
- Health Management Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Jing Wang
- Health Management Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Lian-Sheng Zhang
- Health Management Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| |
Collapse
|
16
|
He W, Cui L, Zhang C, Zhang X, He J, Xie Y, Chen Y. Sonic hedgehog promotes neurite outgrowth of cortical neurons under oxidative stress: Involving of mitochondria and energy metabolism. Exp Cell Res 2017; 350:83-90. [DOI: 10.1016/j.yexcr.2016.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/06/2016] [Accepted: 11/12/2016] [Indexed: 12/29/2022]
|
17
|
Xue J, Zhang X, Zhang C, Kang N, Liu X, Yu J, Zhang N, Wang H, Zhang L, Chen R, Cui L, Wang L, Wang X. Protective effect of Naoxintong against cerebral ischemia reperfusion injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2016; 182:181-189. [PMID: 26902830 DOI: 10.1016/j.jep.2016.02.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 01/05/2016] [Accepted: 02/14/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Naoxintong (NXT), a renowned traditional Chinese medicine in China, has been used for the treatment of acute and chronic cardio-cerebrovascular diseases in clinic for more than 20 years. AIM OF THE STUDY To evaluate the potential neuroprotective effect of NXT against ischemia reperfusion (I/R) injury in mice and investigate the underlying mechanisms. MATERIALS AND METHODS Focal cerebral I/R injury in adult male CD-1 mice was induced by transient middle cerebral artery occlusion (tMCAO) for 1h followed by reperfusion for 23h. Mice were randomly divided into five groups: Sham group; tMCAO group; Vehicle group; NXT-treated groups at doses of 0.36g/kg and 0.54g/kg. The effects of NXT on murine neurological function were estimated by neurological defect scores, infarct volume and brain water content at 24h after tMCAO. Immunohistochemistry and Western blot were used to detect the expression of LOX-1, pERK1/2 and NF-κB at 24h after tMCAO. qRT-PCR was used to detect the expression of LOX-1 and NF-κB at 24h after tMCAO. RESULTS Compared with Vehicle group, 0.54g/kg group of NXT significantly ameliorated neurological outcome, infarction volume and brain water content, decreased the expression of LOX-1, pERK1/2 and NF-κB (P<0.05). CONCLUSION NXT protected the mice brain against I/R injury, and this protection maybe associated with the down-regulation of LOX-1, pERK1/2 and NF-κB expression.
Collapse
Affiliation(s)
- Jing Xue
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei, 050000, PR China.
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China
| | - Ning Kang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China
| | - Xiaoxia Liu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China
| | - Jingying Yu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China
| | - Nan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China
| | - Hong Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China
| | - Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China
| | - Rong Chen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei, 050000, PR China
| | - Lili Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei, 050000, PR China
| | - Lina Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei, 050000, PR China
| | - Xiaolu Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei, 050000, PR China
| |
Collapse
|
18
|
Zhang L, Zhang X, Zhang C, Bai X, Zhang J, Zhao X, Chen L, Wang L, Zhu C, Cui L, Chen R, Zhao T, Zhao Y. Nobiletin promotes antioxidant and anti-inflammatory responses and elicits protection against ischemic stroke in vivo. Brain Res 2016; 1636:130-141. [PMID: 26874072 DOI: 10.1016/j.brainres.2016.02.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Post-ischemic oxidative stress and inflammation play pivotal roles in the pathogenesis of ischemic stroke and may represent therapeutic targets. Nobiletin (NOB) has been reported to elicit a variety of biological effects through its anti-oxidant and anti-inflammatory properties. Our previous study has demonstrated the beneficial effect of NOB in ischemic stroke, but the underlying mechanisms remain poorly defined. We therefore further investigated the role of NOB in cerebral ischemia and its potential mechanisms. METHODS Adult male Sprague-Dawley rats were randomly assigned to five groups: Sham (sham-operated+0.05% Tween-80), permanent middle cerebral artery occlusion (pMCAO+0.9% saline), Vehicle (pMCAO+0.05% Tween-80), NOB-L (pMCAO+NOB 10mg/kg) and NOB-H (pMCAO+NOB 25mg/kg) groups. Rats were pre-administered intraperitoneally once daily for 3 days prior to ischemia and then received once again immediately after surgery. Neurological deficit, brain edema and infarct volume were evaluated at 24h after stroke. Immunohistochemistry, western blot and RT-qPCR were used to detect the expression of Nrf2, HO-1, SOD1, NF-κB and MMP-9. SOD1, GSH and MDA were measured by spectrophotometer. RESULTS Compared with Vehicle group, neurological deficits and brain edema were relieved in NOB-H group, infarct volume was lessened in both NOB-L and NOB-H groups (P<0.05). NOB significantly increased the expression of Nrf2, HO-1, SOD1 and GSH, while decreased the levels of NF-κB, MMP-9 and MDA (P<0.05). CONCLUSION NOB may have a neuroprotective effect on cerebral ischemia, and this protection may be through upregulating Nrf2, HO-1 and downregulating NF-κB expression.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China.
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Xue Bai
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Jian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Xumeng Zhao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Linyu Chen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Lina Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Chunhua Zhu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Lili Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Rong Chen
- Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| | - Ting Zhao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Yuan Zhao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, PR China
| |
Collapse
|
19
|
Li P, Su L, Li X, Di W, Zhang X, Zhang C, He T, Zhu X, Zhang Y, Li Y. Remote limb ischemic postconditioning protects mouse brain against cerebral ischemia/reperfusion injury via upregulating expression of Nrf2, HO-1 and NQO-1 in mice. Int J Neurosci 2015; 126:552-559. [DOI: 10.3109/00207454.2015.1042973] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
20
|
Hu Y, Duan M, Liang S, Wang Y, Feng Y. Senkyunolide I protects rat brain against focal cerebral ischemia–reperfusion injury by up-regulating p-Erk1/2, Nrf2/HO-1 and inhibiting caspase 3. Brain Res 2015; 1605:39-48. [DOI: 10.1016/j.brainres.2015.02.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 02/06/2015] [Accepted: 02/08/2015] [Indexed: 10/24/2022]
|
21
|
SOLICHOVÁ D, BLÁHA M, AUFARTOVÁ J, KRCMOVÁ LK, PLÍŠEK J, HONEGROVÁ B, KASALOVÁ E, LÁNSKÁ M, URBÁNEK L, SOBOTKA L. The Effect of LDL-Apheresis and Rheohaemapheresis Treatment on Vitamin E. J Nutr Sci Vitaminol (Tokyo) 2015; 61:105-12. [DOI: 10.3177/jnsv.61.105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Dagmar SOLICHOVÁ
- 3rd Internal Gerontometabolic Clinic, University Hospital Hradec Králové and Charles University in Prague, Faculty of Medicine in Hradec Králové
| | - Milan BLÁHA
- 4th Internal Clinic-Haematology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové
| | - Jana AUFARTOVÁ
- Department of Analytical Chemistry, Charles University in Prague, Faculty of Pharmacy in Hradec Králové
- 3rd Internal Gerontometabolic Clinic, University Hospital Hradec Králové and Charles University in Prague, Faculty of Medicine in Hradec Králové
| | - Lenka Kujovská KRCMOVÁ
- Department of Analytical Chemistry, Charles University in Prague, Faculty of Pharmacy in Hradec Králové
- 3rd Internal Gerontometabolic Clinic, University Hospital Hradec Králové and Charles University in Prague, Faculty of Medicine in Hradec Králové
| | - Jirí PLÍŠEK
- Department of Analytical Chemistry, Charles University in Prague, Faculty of Pharmacy in Hradec Králové
- 3rd Internal Gerontometabolic Clinic, University Hospital Hradec Králové and Charles University in Prague, Faculty of Medicine in Hradec Králové
| | - Barbora HONEGROVÁ
- Department of Analytical Chemistry, Charles University in Prague, Faculty of Pharmacy in Hradec Králové
| | - Eva KASALOVÁ
- Department of Analytical Chemistry, Charles University in Prague, Faculty of Pharmacy in Hradec Králové
- 3rd Internal Gerontometabolic Clinic, University Hospital Hradec Králové and Charles University in Prague, Faculty of Medicine in Hradec Králové
| | - Miriam LÁNSKÁ
- 4th Internal Clinic-Haematology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové
| | - Lubor URBÁNEK
- Laboratory of Growth Regulators, Faculty of Sciences, Palacky University & Institute of Experimental Botany
| | - Luboš SOBOTKA
- 3rd Internal Gerontometabolic Clinic, University Hospital Hradec Králové and Charles University in Prague, Faculty of Medicine in Hradec Králové
| |
Collapse
|
22
|
Zhang J, Fu B, Zhang X, Zhang L, Bai X, Zhao X, Chen L, Cui L, Zhu C, Wang L, Zhao Y, Zhao T, Wang X. Bicyclol upregulates transcription factor Nrf2, HO-1 expression and protects rat brains against focal ischemia. Brain Res Bull 2013; 100:38-43. [PMID: 24252362 DOI: 10.1016/j.brainresbull.2013.11.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 10/30/2013] [Accepted: 11/06/2013] [Indexed: 01/27/2023]
Abstract
UNLABELLED Oxidative damage plays a detrimental role in the pathophysiology of cerebral ischemia and may represent a therapeutic target. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) controls the coordinated expression of the important antioxidant and detoxification genes through a promotor sequence termed the antioxidant response element. Bicyclol has been proved to elicit a variety of biological effects through its antioxidant and anti-inflammatory properties. But the underlying mechanisms are poorly understood. In this study, the role of bicyclol in cerebral ischemia and its potential mechanism were investigated. METHODS Male Sprague-Dawley rats were randomly assigned to five groups: MCAO (middle cerebral artery occlusion), Vehicle (MCAO+0.5% sodium carboxymethylcellulose), By-L (Vehicle+bicyclol 50mg/kg), By-H (Vehicle+bicyclol 100mg/kg) and Sham operated groups. Bicyclol was administered intragastrically once a day for 3 consecutive days; after 1h of bicyclol pretreatment on the third day, rat ischemic stroke was induced by MCAO. Neurological deficit, infarct volume, and brain edema were detected at 24h after stroke. Western blot and RT-qPCR were used to measure the expression of Nrf2, HO-1 and SOD1. MDA was detected by the spectrophotometer. RESULTS Compared with MCAO group, By-H group significantly ameliorated neurological deficit, lessened the infarct volume and brain edema, increased the expression of Nrf2, HO-1 and SOD1 (P<0.05), and decreased the content of MDA (P<0.05). CONCLUSIONS Bicyclol protected the rat brain from ischemic damage caused by MCAO, and this effect may be through the upregulation of the transcription factor Nrf2 expression.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Baosheng Fu
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China; Hebei Key Laboratory for Neurology, Shijiazhuang, Hebei 050000, PR China.
| | - Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xue Bai
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xumeng Zhao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Linyu Chen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Lili Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Chunhua Zhu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Lina Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Yuan Zhao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Ting Zhao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xiaolu Wang
- Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, PR China
| |
Collapse
|
23
|
Metabonomic strategy to the evaluation of chinese medicine compound danshen dripping pills interfering myocardial ischemia in rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:718305. [PMID: 23737844 PMCID: PMC3659432 DOI: 10.1155/2013/718305] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 03/17/2013] [Accepted: 03/29/2013] [Indexed: 11/18/2022]
Abstract
Coronary heart disease (CHD) is one of the highest mortality diseases in the world. Traditional Chinese medicine compound Danshen dripping pills (CDDPs) have currently made a great achievement in treating CHD. However, the therapeutic mechanism of CDDP is often poorly interpreted. In this study, a GC-MS-based metabonomic study was conducted to assess the holistic efficacy of CDDP for myocardial infarction in male Sprague-Dawley rats, which were divided into the control group, the sham group, the model group, the control + CDDP group, and the model + CDDP, with CDDP at a dose of 107 mg/kg·d (equal to 1.8 mL/kg·d). The metabonomic findings demonstrated great differences of metabolic pattern among sham, model, and the model + CDDP in the orthogonal partial least squares discriminant analysis (OPLS-DA) models, which coordinated well with the assessment of plasma biochemistry and histopathological assay. Differentially expressed metabolites suggested that energy metabolism, glycolysis, and lipid metabolism might be disrupted by myocardial infarction. Both the potential metabolic biomarkers and the biochemical histopathological indices were regulated effectively by CDDP.
Collapse
|
24
|
Li ZL, Hu J, Li YL, Xue F, Zhang L, Xie JQ, Liu ZH, Li H, Yi DH, Liu JC, Wang SW. The effect of hyperoside on the functional recovery of the ischemic/reperfused isolated rat heart: potential involvement of the extracellular signal-regulated kinase 1/2 signaling pathway. Free Radic Biol Med 2013; 57:132-40. [PMID: 23291593 DOI: 10.1016/j.freeradbiomed.2012.12.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 11/21/2012] [Accepted: 12/21/2012] [Indexed: 01/22/2023]
Abstract
One of the leading causes of death in the world is ischemia/reperfusion (I/R)-mediated acute myocardial infarction. There are a lot of Chinese traditional patent medicines, such as Xin'an capsules, Xin Xuening tablets, and so on, which have protective effects against myocardial I/R injury and have been routinely used in treating cardiac diseases for a long time in China. Hyperoside (Hyp) is the chief component of these medicines. This study investigated the action of Hyp in isolated myocardial I/R injury, as well as its possible mechanisms. Using the Langendorff model, isolated Sprague-Dawley rat hearts were subjected to 30 min of global ischemia and 50 min of reperfusion. Cardiac function was measured, and infarct size was evaluated by triphenyltetrazolium chloride staining at the end of the reperfusion. Coronary effluent was analyzed for lactate dehydrogenase (LDH) and creatine kinase (CK). Myocardium was also measured for total superoxide dismutase (SOD) activity and malondialdehyde (MDA) content. Phosphorylation of extracellular signal-regulated protein kinase (ERK) was analyzed by Western blotting. We report for the first time that administration of Hyp before/after I/R significantly improved heart contraction and limited the infarct size and CK and LDH leakage from the damaged myocardium after I/R. The activity of SOD and the MDA content remarkably changed in the presence of Hyp as well. Phosphorylation of ERK was significantly increased in Hyp-treated hearts compared to controls (p<0.01). Hyp-induced ERK phosphorylation was inhibited by PD98059. We therefore conclude that Hyp can protect cardiomyocytes from I/R-induced oxidative stress through the activation of ERK-dependent signaling.
Collapse
Affiliation(s)
- Zi-lin Li
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Neumann CL, Schulz EG, Hagenah GC, Platzer U, Wieland E, Schettler V. Lipoprotein apheresis – More than just cholesterol reduction? ATHEROSCLEROSIS SUPP 2013; 14:29-32. [DOI: 10.1016/j.atherosclerosissup.2012.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Chen L, Wang L, Zhang X, Cui L, Xing Y, Dong L, Liu Z, Li Y, Zhang X, Wang C, Bai X, Zhang J, Zhang L, Zhao X. The protection by octreotide against experimental ischemic stroke: up-regulated transcription factor Nrf2, HO-1 and down-regulated NF-κB expression. Brain Res 2012; 1475:80-7. [PMID: 22885292 DOI: 10.1016/j.brainres.2012.07.052] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/24/2012] [Accepted: 07/25/2012] [Indexed: 12/25/2022]
Abstract
BACKGROUND Inflammatory and oxidative damage play a pivotal role in cerebral ischemic pathogenesis and may represent a therapeutic target. Octreotide (OCT) has been proved to elicit a variety of biological effects through its anti-inflammatory and anti-oxidant properties in the treatment of severe acute pancreatitis and ischemia-reperfusion injury in retina and intestine. However little is known regarding the effect of OCT in ischemic stroke. Here, we designed this study to investigate the protective effect of OCT in ischemic stroke and explore the potential underlying mechanisms. METHODS Male Sprague-Dawley rats were subjected to permanent middle cerebral artery occlusion (pMCAO) and randomly divided into four groups: Sham (sham-operated), MCAO (pMCAO+0.9% saline), OCT-L (pMCAO+OCT 50μg/kg) and OCT-H (pMCAO+OCT 100μg/kg) groups. OCT was administered intraperitoneally immediately after stroke. Neurological deficit scores, infarct volume and brain water content were measured at 24h after stroke. Immunohistochemical staining and western blot were used to analyze the expressions of Nrf2, HO-1 and NF-κB. SOD and MDA were measured by spectrophotometer. RESULTS Compared with MCAO group, OCT significantly alleviated neurological deficit, lessened infarct volume and brain edema (P<0.05), upregulated the expression of Nrf2, HO-1 and SOD (P<0.05), and decreased the expression of NF-κB and MDA (P<0.05). CONCLUSIONS OCT protected the brain against cerebral ischemic damage; this effect may be through upregulation of transcription factor Nrf2, HO-1 and downregulation of NF-κB expression.
Collapse
Affiliation(s)
- Linyu Chen
- Department of Neurology, Second Hospital of Hebei Medical University; Shijiazhuang, Hebei 050000, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lu X, Rasco BA. Determination of Antioxidant Content and Antioxidant Activity in Foods using Infrared Spectroscopy and Chemometrics: A Review. Crit Rev Food Sci Nutr 2012; 52:853-75. [DOI: 10.1080/10408398.2010.511322] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
28
|
Protective Effect of Luteolin in Experimental Ischemic Stroke: Upregulated SOD1, CAT, Bcl-2 and Claudin-5, Down-Regulated MDA and Bax Expression. Neurochem Res 2012; 37:2014-24. [DOI: 10.1007/s11064-012-0822-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 05/29/2012] [Accepted: 05/31/2012] [Indexed: 12/17/2022]
|
29
|
Chen TS, Liou SY, Wu HC, Tsai FJ, Tsai CH, Huang CY, Chang YL. Low-density lipoprotein (LDL) apheresis reduces atherogenic and oxidative markers in uremic patients with hyperlipidemia. Int Urol Nephrol 2010; 43:471-4. [DOI: 10.1007/s11255-010-9722-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 02/23/2010] [Indexed: 11/27/2022]
|
30
|
Passauer J, Herbrig K, Fischer S, Bornstein S, Julius U. A single lipid apheresis does not modulate pulse wave reflection in hypercholesterolemic patients. ATHEROSCLEROSIS SUPP 2009; 10:44-8. [DOI: 10.1016/s1567-5688(09)71809-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
31
|
Wei Y, Zhang R. Preventive Effect of Fastigial Nucleus on Oxidative Damage in Rats Undergoing Acute Myocardial Infarction. ACTA ACUST UNITED AC 2008. [DOI: 10.1248/jhs.54.330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yidong Wei
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University
| | - Runfeng Zhang
- Heart, Lung and Blood Vessel Center, Tongji University
| |
Collapse
|
32
|
Bugeja AL, Chan CT. Improvement in Lipid Profile by Nocturnal Hemodialysis in Patients with End-Stage Renal Disease. ASAIO J 2004; 50:328-31. [PMID: 15307542 DOI: 10.1097/01.mat.0000130518.62960.43] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Dyslipidemia is associated with uremia and an increased risk of cardiovascular disease. The uremic dyslipidemia syndrome is characterized by an abnormal lipoprotein profile that results in (1) an elevation of triglyceride (TG) rich lipoproteins, very low density lipoprotein (VLDL), and intermediate density lipoprotein (IDL); (2) a reduction in high density lipoprotein (HDL) levels; and (3) a higher fraction of atherogenic, small dense low density lipoprotein (LDL). Nocturnal hemodialysis (NHD) is a home based renal replacement therapy that provides better control of uremia than conventional hemodialysis (CHD) and that may improve dyslipidemia. To test this hypothesis, we conducted a prospective cohort study of 11 patients with end-stage renal disease (ESRD) (age 38+/-3 years [mean+/-SEMI) before and after conversion from CHD to NHD. Weight, blood pressure (BP), serum hemoglobin (Hb), phosphate (PO4), and albumin (Alb) were assessed at baseline and at 3 months after conversion to NHD. Dialysis dose on CHD and NHD was assessed using equilibrated Kt/V (eKt/V). A 12 hour fasting lipid profile (total cholesterol [TC], TG, HDL, LDL, HDL/TC) was obtained once while on CHD and at 3 months after conversion to NHD. After conversion from CHD to NHD, eKt/V per session increased significantly (from 1.13+/-0.05 to 2.10+/-0.07; p < 0.05). TG level decreased significantly (from 2.05+/-0.30 to 1.01+/-0.14 mmol/L; p < 0.001), and HDL level increased significantly (from 1.17+/-0.13 to 1.65+/-0.14 mmol/L; p < 0.001). HDL/TC also increased significantly (from 0.26+/-0.03 to 0.35+/-0.02; p < 0.001). TC and LDL levels were unchanged. HDL levels increased and TG levels decreased in all patients. There was no difference in weight, Hb, and Alb. Systolic BP and PO4 were significantly lower, and there was a trend toward a reduction in cardiovascular medications. The mechanism for the improvement in lipid profile requires further study.
Collapse
Affiliation(s)
- Ann L Bugeja
- Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
33
|
Malliaraki N, Mpliamplias D, Kampa M, Perakis K, Margioris AN, Castanas E. Total and corrected antioxidant capacity in hemodialyzed patients. BMC Nephrol 2003; 4:4. [PMID: 12837136 PMCID: PMC166281 DOI: 10.1186/1471-2369-4-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2003] [Accepted: 07/01/2003] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Oxidative stress may play a critical role in the vascular disease of end stage renal failure and hemodialysis patients. Studies, analyzing either discrete analytes and antioxidant substances, or the integrated total antioxidant activity of human plasma during hemodialysis, give contradictory results. METHODS Recently, we have introduced a new automated method for the determination of Total Antioxidant Capacity (TAC) of human plasma. We have serially measured TAC and corrected TAC (cTAC: after subtraction of the interactions due to endogenous uric acid, bilirubin and albumin) in 10 patients before the onset of the dialysis session, 10 min, 30 min, 1 h, 2 h and 3 h into the procedure and after completion of the session. RESULTS Our results indicate that TAC decreases, reaching minimum levels at 2 h. However, corrected TAC increases with t1/2 of about 30 min. We then repeated the measurements in 65 patients undergoing dialysis with different filters (36 patients with ethylene vinyl alcohol copolymer resin filter -Eval-, 23 patients with two polysulfone filters -10 with F6 and 13 with PSN140-, and 6 patients with hemophan filters). Three specimens were collected (0, 30, 240 min). The results of this second group confirm our initial results, while no significant difference was observed using either filter. CONCLUSIONS Our results are discussed under the point of view of possible mechanisms of modification of endogenous antioxidants, and the interaction of lipid- and water-soluble antioxidants.
Collapse
Affiliation(s)
- Niki Malliaraki
- Departments of Clinical Chemistry, University of Crete, School of Medicine, and University Hospital, Heraklion, GR-71110, Greece
| | - Dimitris Mpliamplias
- Experimental Endocrinology, University of Crete, School of Medicine, and University Hospital, Heraklion, GR-71110, Greece
| | - Marilena Kampa
- Experimental Endocrinology, University of Crete, School of Medicine, and University Hospital, Heraklion, GR-71110, Greece
| | - Kostas Perakis
- Nephrology, University of Crete, School of Medicine, and University Hospital, Heraklion, GR-71110, Greece
| | - Andrew N Margioris
- Departments of Clinical Chemistry, University of Crete, School of Medicine, and University Hospital, Heraklion, GR-71110, Greece
| | - Elias Castanas
- Experimental Endocrinology, University of Crete, School of Medicine, and University Hospital, Heraklion, GR-71110, Greece
| |
Collapse
|
34
|
Schettler V, Krontal J, Scheel A, Wieland E. No acute impact of lipid apheresis treatment on free radical scavenging enzyme gene expression in white blood cells. Eur J Clin Invest 2003; 33:134-40. [PMID: 12588287 DOI: 10.1046/j.1365-2362.2003.01119.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Lipid apheresis (LA) treatment has been suggested to cause oxidative stress. Defense against oxygen-radical-mediated damage is provided by nonenzymatic and enzymatic antioxidants. In the present investigation we have investigated whether gene expression of free radical scavenging enzymes (FRSE) is affected in leukocytes of patients undergoing LDL-apheresis. MATERIALS AND METHODS For this purpose cellular glutathione peroxidase (GPx-1), phospholipid glutathione peroxidase (GPx-4), glutathione reductase (GSSG-R), glutathione synthetase (GSH-S), Cu/Zn-superoxide dismutase (SOD-1) and catalase (CAT) mRNA expression were followed at the start (SA) and immediately after (EA) LA treatment (n = 25). Gene expression was determined by quantitative RT-PCR with the LightCycler(R) instrument (Roche Diagnostics, Mannheim, Germany) and transcription elongation factor-2 as reference gene. RESULTS The expression of GPx-1, GPx-4, GSSG-R, GSH-S, SOD-1, CAT mRNA was not affected by a single LA treatment. Free radical scavenging enzymes mRNAs were significantly (P < 0.05) increased in the LA patients (GPx-1: 2.00 +/- 1.37; GPx-4: 0.52 +/- 0.46; GSSG-R: 0.07 +/- 0.03; GSH-S: 0.04 +/- 0.03; SOD-1: 1.12 +/- 0.74; CAT: 0.15 +/- 0.07) when compared with 26 healthy blood donors (GPx-1: 1.1 +/- 0.6; GPx-4: 0.35 +/- 0.19; GSSG-R: 0.02 +/- 0.01; GSH-S: 0.03 +/- 0.01; SOD-1: 0.16 +/- 0.08; CAT: 0.09 +/- 0.05; mean +/- SD). CONCLUSIONS These results show that the LA procedure does not acutely affect the antioxidant defense system on the gene level but suggests that the chronic stress resulting from hyperlipidaemia and/or LA may cause FRSE gene induction.
Collapse
Affiliation(s)
- V Schettler
- Department of Nephrology and Rheumatology, Georg-August University, Göttingen, Germany.
| | | | | | | |
Collapse
|
35
|
Napoli C, Lerman LO. Involvement of oxidation-sensitive mechanisms in the cardiovascular effects of hypercholesterolemia. Mayo Clin Proc 2001; 76:619-31. [PMID: 11393501 DOI: 10.4065/76.6.619] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Hypercholesterolemia is a common clinical metabolic and/or genetic disorder that promotes functional and structural vascular wall injury. The underlying mechanisms for these deleterious effects involve a local inflammatory response and release of cytokines and growth factors. Consequent activation of oxidation-sensitive mechanisms in the arterial wall, modulation of intracellular signaling pathways, increased oxidation of low-density lipoprotein cholesterol, and quenching of nitric oxide can all impair the functions controlled by the vascular wall and lead to the development of atherosclerosis. This cascade represents a common pathological mechanism activated by various cardiovascular risk factors and may partly underlie synergism among them as well as the early pathogenesis of atherosclerosis. Antioxidant intervention and restoration of the bioavailability of nitric oxide have been shown to mitigate functional and structural arterial alterations and improve cardiovascular outcomes. Elucidation of the precise nature and role of early transductional signaling pathways and transcriptional events activated in hypercholesterolemia in children and adults, including mothers during pregnancy, and understanding their downstream effects responsible for atherogenesis may help in directing preventive and interventional measures against atherogenesis and vascular dysfunction.
Collapse
Affiliation(s)
- C Napoli
- Department of Medicine, University of Naples, Italy.
| | | |
Collapse
|