1
|
Chao HWH, Chao WWJ, Chao HM. Catalpol Protects Against Retinal Ischemia Through Antioxidation, Anti-Ischemia, Downregulation of β-Catenin, VEGF, and Angiopoietin-2: In Vitro and In Vivo Studies. Int J Mol Sci 2025; 26:4019. [PMID: 40362263 PMCID: PMC12072090 DOI: 10.3390/ijms26094019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Retinal ischemic disorders present significant threats to vision, characterized by inadequate blood supply oxygen-glucose deprivation (OGD), oxidative stress, and cellular injury, often resulting in irreversible injury. Catalpol, an iridoid glycoside derived from Rehmannia glutinosa, has demonstrated antioxidative and neuroprotective effects. This study aimed at investigating the protective effects and mechanisms of catalpol against oxidative stress or OGD in vitro and retinal ischemia in vivo, focusing on the modulation of key biomarkers of retinal ischemia, including HIF-1α, vascular endothelial growth factor (VEGF), angiopoietin-2, MCP-1, and the Wnt/β-catenin pathway. Cellular viability was assessed using retinal ganglion cell-5 (RGC-5) cells cultured in DMEM; a 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed. H2O2 (1 mM)/OGD was utilized. Vehicle or different catalpol concentrations were administered 15 min before the ischemic-like insults. The Wistar rat eyes' intraocular pressure was increased to 120 mmHg for 60 min to induce retinal ischemia. Intravitreous injections of catalpol (0.5 or 0.25 mM), Wnt inhibitor DKK1 (1 μg/4 μL), anti-VEGF Lucentis (40 μg/4 μL), or anti-VEGF Eylea (160 μg/4 μL) were administered to the rats' eyes 15 min before or after retinal ischemia. Electroretinogram (ERG), fluorogold retrograde labeling RGC, Western blotting, ELISA, RT-PCR, and TUNEL were utilized. In vitro, both H2O2 and OGD models significantly (p < 0.001/p < 0.001; H2O2 and OGD) induced oxidative stress/ischemic-like insults, decreasing RGC-5 cell viability (from 100% to 55.14 ± 2.19%/60.84 ± 4.57%). These injuries were insignificantly (53.85 ± 1.28% at 0.25 mM)/(63.46 ± 3.30% at 0.25 mM) and significantly (p = 0.003/p = 0.012; 64.15 ± 2.41%/77.63 ± 8.59% at 0.5 mM) altered by the pre-administration of catalpol, indicating a possible antioxidative and anti-ischemic effect of 0.5 mM catalpol. In vivo, catalpol had less effect at 0.25 mM for ERG amplitude ratio (median [Q1, Q3] 14.75% [12.64%, 20.48%]) and RGC viability (mean ± SE 63.74 ± 5.13%), whereas (p < 0.05 and p < 0.05) at 0.5 mM ERG's ratio (35.43% [24.35%, 43.08%]) and RGC's density (74.34 ± 5.10%) blunted the ischemia-associated significant (p < 0.05 and p < 0.01) reduction in ERG b-wave amplitude (6.89% [4.24%, 10.40%]) and RGC cell viability (45.64 ± 3.02%). Catalpol 0.5 mM also significantly protected against retinal ischemia supported by the increased amplitude ratio of ERG a-wave and oscillatory potential, along with recovering a delayed a-/b-wave response time ratio. When contrasted with DKK1 or Lucentis, catalpol exhibited similar protective effects against retinal ischemia via significantly (p < 0.05) blunting the ischemia-induced overexpression of β-catenin, VEGF, or angiopoietin-2. Moreover, ischemia-associated significant increases in apoptotic cells in the inner retina, inflammatory biomarker MCP-1, and ischemic indicator HIF-1α were significantly nullified by catalpol. Catalpol demonstrated antiapoptotic, anti-inflammatory, anti-ischemic (in vivo retinal ischemia or in vitro OGD), and antioxidative (in vitro) properties, counteracting retinal ischemia via suppressing upstream Wnt/β-catenin and inhibiting downstream HIF-1α, VEGF, and angiopoietin-2, together with its decreasing TUNEL apoptotic cell number and inflammatory MCP-1 concentration.
Collapse
Affiliation(s)
- Howard Wen-Haur Chao
- Department of Medicine, School of Medicine, Aston University, Birmingham B4 7ET, UK
- Department of Medical Education, Leeds University, Leeds LS2 9JT, UK
| | - Windsor Wen-Jin Chao
- Department of Medicine, School of Medicine, Aston University, Birmingham B4 7ET, UK
- Department of Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Hsiao-Ming Chao
- Department of Chinese Medicine, School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Institute of Pharmacology, Department of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Department of Ophthalmology, Shin Kong Hospital, Taipei 105, Taiwan
| |
Collapse
|
2
|
Cairang N, Wu Y, Zhi S, Tang J, Tie X, Zhan D, Lu G, Shi Y, Zhao Q. 5-(3-( N-(Carboxymethyl)naphthalene-2-sulfonamido)phenyl)-1-ethyl-1 H-pyrrole-2-carboxylic acid as a Keap1-Nrf2 inhibitor for cerebral ischemia/reperfusion injury treatment. RSC Adv 2025; 15:1052-1059. [PMID: 39807188 PMCID: PMC11726444 DOI: 10.1039/d4ra06512c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/23/2024] [Indexed: 01/16/2025] Open
Abstract
The Keap1 (Kelch-like ECH-Associating Protein 1)-Nrf2 (Nuclear Factor Erythroid 2-Related Factor 2)-ARE (Antioxidant Response Element) signaling pathway plays a crucial role in the oxidative stress response and has been linked to the development and progression of various diseases. Its influence on cerebral ischemia/reperfusion (I/R) injury has garnered significant attention. In our study, we investigated the effect of compound 2, a non-covalent inhibitor of the Keap1-Nrf2 interaction, which was previously discovered by our research group. Specifically, we used 5-(3-(N-(carboxymethyl)naphthalene-2-sulfonamido)phenyl)-1-ethyl-1H-pyrrole-2-carboxylic acid (compound 2) to assess its therapeutic potential in a cerebral I/R injury model. The results demonstrated that compound 2 had a significant therapeutic effect, promoting the translocation of Nrf2 from the cytoplasm to the nucleus in diseased tissue. Additionally, it increased the production of key antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH).
Collapse
Affiliation(s)
- Nanjia Cairang
- Department of Tibetan Medicine, University of Xizang Medicine 10 Dangre Middle Road, Chengguan District Lhasa City 850000 China
| | - Yanran Wu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University 1160 Shengli Street Yinchuan 750004 China
| | - Shumeng Zhi
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University 1160 Shengli Street Yinchuan 750004 China
| | - Jiaqin Tang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University 1160 Shengli Street Yinchuan 750004 China
| | - Xin Tie
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University 1160 Shengli Street Yinchuan 750004 China
| | - Dui Zhan
- Department of Tibetan Medicine, University of Xizang Medicine 10 Dangre Middle Road, Chengguan District Lhasa City 850000 China
| | - Guangyuan Lu
- Ningxia Key Laboratory of Craniocerebral Diseases, Incubation Base of National Key Laboratory, Ningxia Medical University 1160 Shengli Street Yinchuan 750004 China
| | - Ying Shi
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University 1160 Shengli Street Yinchuan 750004 China
| | - Qipeng Zhao
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area (Ningxia Medical University), Ministry of Education, School of Pharmacy, Ningxia Medical University 1160 Shengli Street Yinchuan 750004 China
| |
Collapse
|
3
|
Chen H, Deng C, Meng Z, Zhu M, Yang R, Yuan J, Meng S. Combined Catalpol and Tetramethylpyrazine Promote Axonal Plasticity in Alzheimer's Disease by Inducing Astrocytes to Secrete Exosomes Carrying CDK5 mRNA and Regulating STAT3 Phosphorylation. Mol Neurobiol 2024; 61:10770-10791. [PMID: 38789892 DOI: 10.1007/s12035-024-04251-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Alzheimer's disease (AD) is a common progressive degenerative disease of the central nervous system in aging populations. This study aimed to investigate the effects of combined catalpol and tetramethylpyrazine (CT) in promoting axonal plasticity in AD and the potential underlying mechanism. Astrocytes were treated with different concentrations of compatible CT. Exosomes were collected and subjected to sequencing analysis, which was followed by the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of differentially expressed genes. Amyloid precursor protein/presenilin 1 (APP/PS1) double-transfected male mice were used as the in vivo AD models. Astrocyte-derived exosomes that were transfected with cyclin-dependent kinase 5 (CDK5) or CT treatment were injected into the tail vein of mice. The levels of CDK5, synaptic plasticity marker protein neurofilament 200 (NF200), and growth-associated protein 43 (GAP-43) in the hippocampus of mice were compared in each group. Immunofluorescence staining was used to detect the localization of STAT3 and to visualize synaptic morphology via β-tubulin-III (TUBB3). Astrocyte-derived exosomes transfected with siCDK5 or treated with CT were co-cultured with HT-22 cells, which were untransfected or silenced for signal transducer and activator of transcription 3 (STAT3). Amyloid β-protein (Aβ)1-42 was induced in the in vitro AD models. The viability, apoptosis, and expression levels of NF200 and GAP-43 proteins in the hippocampal neurons of each group were compared. In total, 166 differentially expressed genes in CT-induced astrocyte-derived exosomes were included in the KEGG analysis, and they were found to be enriched in 12 pathways, mainly in axon guidance. CT treatment significantly increased the level of CDK5 mRNA in astrocyte-derived exosomes-these exosomes restored CDK5 mRNA and protein levels in the hippocampus of the in vivo AD model mice and the in vitro AD model; promoted p-STAT3 (Ser727), NF200 and GAP-43 proteins; and promoted the regeneration and extension of neuronal synapses. Silencing of CDK5 blocked both neuronal protection as well as induction of axonal plasticity in AD by CT-treated exosomes in vitro and in vivo. Moreover, silencing of STAT3 blocked both neuronal protection as well as induction of axonal plasticity in AD caused by CDK5 overexpression or CT-treated astrocyte-induced exosomes. CT promotes axonal plasticity in AD by inducing astrocytes to secrete exosomes carrying CDK5 mRNA and regulating STAT3 (Ser727) phosphorylation.
Collapse
Affiliation(s)
- Huize Chen
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,Xuhui District, 600 Yishan Road, Shanghai, 200233, China
| | - Chujun Deng
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,Xuhui District, 600 Yishan Road, Shanghai, 200233, China
| | - Zeyu Meng
- Second Clinical Medicine College of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Mengting Zhu
- Graduate School of Jiangxi, University of Traditional Chinese Medicine, Nanchang, China
| | - Ruoyu Yang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Yuan
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,Xuhui District, 600 Yishan Road, Shanghai, 200233, China
| | - Shengxi Meng
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,Xuhui District, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
4
|
Shen F, Yang W, Luan G, Peng J, Li Z, Gao J, Hou Y, Bai G. Catalpolaglycone disrupts mitochondrial thermogenesis by specifically binding to a conserved lysine residue of UCP2 on the proton leak tunnel. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155356. [PMID: 38241920 DOI: 10.1016/j.phymed.2024.155356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/03/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
BACKGROUND Catalpol (CAT), a naturally occurring iridoid glycoside sourced from the root of Rehmannia glutinosa, affects mitochondrial metabolic functions. However, the mechanism of action of CAT against pyrexia and its plausible targets remain to be fully elucidated. PURPOSE This study aimed to identify the specific targets of CAT for blocking mitochondrial thermogenesis and to unveil the unique biological mechanism of action of the orthogonal binding mode between the hemiacetal group and lysine residue on the target protein in vivo. METHODS Lipopolysaccharide (LPS)/ carbonyl cyanide 3-chlorophenylhydrazone (CCCP)-induced fever models were established to evaluate the potential antipyretic effects of CAT. An alkenyl-modified CAT probe was designed to identify and capture potential targets. Binding capacity was tested using in-gel imaging and a cellular thermal shift assay. The underlying antipyretic mechanisms were explored using biochemical and molecular biological methods. Catalpolaglycone (CA) was coupled with protein profile identification and molecular docking analysis to evaluate and identify its binding mode to UCP2. RESULTS After deglycation of CAT in vivo, the hemiacetal group in CA covalently binds to Lys239 of UCP2 in the mitochondria of the liver via an ɛ-amine nucleophilic addition. This irreversible binding affects proton leakage and improves mitochondrial membrane potential and ADP/ATP transformation efficiency, leading to an antipyretic effect. CONCLUSION Our findings highlight the potential role of CA in modulating UCP2 activity or function within the mitochondria and open new avenues for investigating the therapeutic effects of CA on mitochondrial homeostasis.
Collapse
Affiliation(s)
- Fukui Shen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Wen Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Guoqing Luan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Jiamin Peng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Zhenqiang Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| |
Collapse
|
5
|
Effects of Catalpol on Alzheimer's Disease and Its Mechanisms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2794243. [PMID: 35815283 PMCID: PMC9262514 DOI: 10.1155/2022/2794243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/17/2022] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system characterized by memory loss and cognitive dysfunction. With the increasing aging of the population, the incidence of AD and the number of patients are also increasing year by year, causing more and more heavy burdens to the family and society. Catalpol, an iridoid glycoside compound, is one of the main active components of Rehmannia glutinosa. At present, a large number of experimental studies in vivo and in vitro have confirmed that catalpol has antioxidant, anti-inflammatory, antiapoptotic, and other neuroprotective effects, and it plays a significant role in the prevention and treatment of AD, with very small side effects and high safety. Therefore, it may be an ideal drug for the treatment of AD. Based on this, the role and mechanism of catalpol in AD will be comprehensively reviewed in the following.
Collapse
|
6
|
Ge H, Lin W, Lou Z, Chen R, Shi H, Zhao Q, Lin Z. Catalpol alleviates myocardial ischemia reperfusion injury by activating the Nrf2/HO-1 signaling pathway. Microvasc Res 2022; 140:104302. [PMID: 34919942 DOI: 10.1016/j.mvr.2021.104302] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 11/15/2022]
Abstract
PURPOSE Myocardial ischemia/reperfusion injury (MI/RI) is a major problem in the clinical treatment of ischemic cardiomyopathy, and its specific underlying mechanisms are complicated and still unclear. A number of studies have indicated that the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxidase-1(HO-1) signaling pathway might serve as an important target for the management of MI/RI. Catalpol is a kind of iridoid glucoside that has been found to exhibit diverse anti-inflammatory and antioxidant properties. This study was aimed at investigating the role of Catalpol in targeting MI/RI and its related mechanisms in an oxygen-glucose deprivation/reoxygenation (OGD/R) model in vitro and a preclinical ischemia/reperfusion (I/R) model. METHODS This study using both in vitro and in vivo models investigated the possible role and underlying mechanisms used by Catalpol for modulating of MI/RI. The potential effects of Catalpol on the viability of cardiomyocytes were measured by cell counting kit-8 (CCK-8) assays. The phenotypes of myocardial injury, oxidative stress and inflammation markers were measured by western blot, immunofluorescence, enzyme-linked immunosorbent assay (ELISA) etc. Nrf2/HO-1 signaling pathway was detected by immunofluorescence and western blot analysis. RESULTS We found that Catalpol significantly suppressed the process of MI/RI and protected OGD/R-treated cardiomyocytes by inhibiting the various markers of inflammation and suppressing oxidative stress. Additionally, mechanistically it was also demonstrated that Catalpol could effectively activate Nrf2/HO-1 signaling pathway to suppress the damage caused by inflammation and oxidative stress in MI/RI. CONCLUSION In summary, the findings suggest that Catalpol exerted significant cardioprotective effects following myocardial ischemia, possibly through the activation of the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Hanwei Ge
- Department of Cardiovascular and Thoracic Surgery, Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wei Lin
- Department of Cardiovascular and Thoracic Surgery. The People's Hospital of Pingyang, Wenzhou, Zhejiang 325400, China
| | - Zhiling Lou
- Department of Cardiovascular and Thoracic Surgery, Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Ruiheng Chen
- Department of Cardiovascular and Thoracic Surgery, Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Haochun Shi
- Department of Cardiovascular and Thoracic Surgery, Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qifeng Zhao
- Department of Cardiovascular and Thoracic Surgery, Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhiyong Lin
- Department of Cardiovascular and Thoracic Surgery, Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
7
|
Zhang X, Liu K, Shi M, Xie L, Deng M, Chen H, Li X. Therapeutic potential of catalpol and geniposide in Alzheimer's and Parkinson's diseases: A snapshot of their underlying mechanisms. Brain Res Bull 2021; 174:281-295. [PMID: 34216649 DOI: 10.1016/j.brainresbull.2021.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/09/2021] [Accepted: 06/29/2021] [Indexed: 01/28/2023]
Abstract
Rehmannia glutinosa, the fresh or dried root of Rehmannia glutinosa (Gaertn.) Libosch. ex Fisch. & Mey., and Gardenia, the fruit of Gardenia jasminoides Ellis from Rubiaceae, both are famous traditional Chinese medicines that have been traditionally used in China. Catalpol and geniposide, as two kinds of iridoid glycosides with high activities, are the main bioactive components in Rehmannia glutinosa and Gardenia jasminoides Ellis, respectively. Over the past few decades, catalpol and geniposide have been widely studied for their therapeutic effects. The preclinical experiments demonstrated that they possessed significant neuroprotective activities against Alzheimer's disease, Parkinson's disease, stroke, and depression, etc. In this paper, the pharmacological effects and mechanisms of catalpol and geniposide on Alzheimer's disease and Parkinson's disease from 2005 to now were systematically summarized and comprehensively analyzed. At the same time, the pharmacokinetic characteristics of the analyzed compounds were also described, hoping to provide some enlightenment for the design, research, and development of iridoid glycosides.
Collapse
Affiliation(s)
- Xumin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Kai Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Mingyi Shi
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Long Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Mao Deng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Huijuan Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
8
|
Xie Q, Li H, Lu D, Yuan J, Ma R, Li J, Ren M, Li Y, Chen H, Wang J, Gong D. Neuroprotective Effect for Cerebral Ischemia by Natural Products: A Review. Front Pharmacol 2021; 12:607412. [PMID: 33967750 PMCID: PMC8102015 DOI: 10.3389/fphar.2021.607412] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Natural products have a significant role in the prevention of disease and boosting of health in humans and animals. Stroke is a disease with high prevalence and incidence, the pathogenesis is a complex cascade reaction. In recent years, it’s reported that a vast number of natural products have demonstrated beneficial effects on stroke worldwide. Natural products have been discovered to modulate activities with multiple targets and signaling pathways to exert neuroprotection via direct or indirect effects on enzymes, such as kinases, regulatory receptors, and proteins. This review provides a comprehensive summary of the established pharmacological effects and multiple target mechanisms of natural products for cerebral ischemic injury in vitro and in vivo preclinical models, and their potential neuro-therapeutic applications. In addition, the biological activity of natural products is closely related to their structure, and the structure-activity relationship of most natural products in neuroprotection is lacking, which should be further explored in future. Overall, we stress on natural products for their role in neuroprotection, and this wide band of pharmacological or biological activities has made them suitable candidates for the treatment of stroke.
Collapse
Affiliation(s)
- Qian Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Danni Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianmei Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinxiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mihong Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Daoyin Gong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
9
|
Network Pharmacology-Based Prediction of Catalpol and Mechanisms against Stroke. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2541316. [PMID: 33505489 PMCID: PMC7810528 DOI: 10.1155/2021/2541316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/19/2020] [Accepted: 12/28/2020] [Indexed: 12/03/2022]
Abstract
Aim To apply the network pharmacology method to screen the target of catalpol prevention and treatment of stroke, and explore the pharmacological mechanism of Catalpol prevention and treatment of stroke. Methods PharmMapper, GeneCards, DAVID, and other databases were used to find key targets. We selected hub protein and catalpol which were screened for molecular docking verification. Based on the results of molecular docking, the ITC was used to determine the binding coefficient between the highest scoring protein and catalpol. The GEO database and ROC curve were used to evaluate the correlation between key targets. Results 27 key targets were obtained by mapping the predicted catalpol-related targets to the disease. Hub genes (ALB, CASP3, MAPK1 (14), MMP9, ACE, KDR, etc.) were obtained in the key target PPI network. The results of KEGG enrichment analysis showed that its signal pathway was involved in angiogenic remodeling such as VEGF, neurotrophic factors, and inflammation. The results of molecular docking showed that ACE had the highest docking score. Therefore, the ITC was used for the titration of ACE and catalpol. The results showed that catalpol had a strong binding force with ACE. Conclusion Network pharmacology combined with molecular docking predicts key genes, proteins, and signaling pathways for catalpol in treating stroke. The strong binding force between catalpol and ACE was obtained by using ITC, and the results of molecular docking were verified to lay the foundation for further research on the effect of catalpol on ACE. ROC results showed that the AUC values of the key targets are all >0.5. This article uses network pharmacology to provide a reference for a more in-depth study of catalpol's mechanism and experimental design.
Collapse
|
10
|
Jiang Y, Zhang Q. Catalpol ameliorates doxorubicin-induced inflammation and oxidative stress in H9C2 cells through PPAR-γ activation. Exp Ther Med 2020; 20:1003-1011. [PMID: 32765656 PMCID: PMC7388568 DOI: 10.3892/etm.2020.8743] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Drug-induced cardiomyopathy is a severe disease that leads to refractory heart disease at late stages, with increasing detrimental effects. DOX-induced cell damage is primarily induced via cellular oxidative stress. The present study investigated the effects of catalpol on doxorubicin (DOX)-induced H9C2 cardiomyocyte inflammation and oxidative stress. The Cell Counting Kit-8 assay was performed to detect cell viability, and western blotting was performed to detect the expression of peroxisome proliferator-activated receptor (PPAR)-γ in H9C2 cells. The expression levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6 were measured using ELISAs. Furthermore, the oxidative stress kit was used to detect the levels of malondialdehyde, superoxide dismutase and glutathione peroxidase. A reactive oxygen species (ROS) kit and DCF-DA staining were used to detect ROS levels. The results indicated that DOX treatment inhibited H9C2 cell expression of PPAR-γ and decreased H9C2 cell viability. Various concentrations of catalpol exhibited a less potent effect on H9C2 cell viability compared with DOX; however, catalpol increased the viability of DOX-induced H9C2 cells. Catalpol treatment also significantly decreased the expression levels of inflammatory factors (TNF-α, IL-1β and IL-6) in DOX-induced H9C2 cells, which was reversed by transfections with short hairpin RNA targeting PPAR-γ. Results from the present study indicated that catalpol ameliorated DOX-induced inflammation and oxidative stress in H9C2 cardiomyoblasts by activating PPAR-γ.
Collapse
Affiliation(s)
- Yanjie Jiang
- Department of Pharmacology, Jinhua Institute for Food and Drug Control, Jinhua, Zhejiang 321017, P.R. China
| | - Qing Zhang
- Department of Pharmacy, Lianshui County People's Hospital, Huai'an, Jiangsu 223400, P.R. China
| |
Collapse
|
11
|
Liu L, Zhao Z, Yin Q, Zhang X. TTB Protects Astrocytes Against Oxygen-Glucose Deprivation/Reoxygenation-Induced Injury via Activation of Nrf2/HO-1 Signaling Pathway. Front Pharmacol 2019; 10:792. [PMID: 31379570 PMCID: PMC6646521 DOI: 10.3389/fphar.2019.00792] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/18/2019] [Indexed: 12/17/2022] Open
Abstract
Neonatal hypoxic/ischemic encephalopathy (NHIE) is a severe condition that leads to death or neurological disability in newborns. The underlying pathological mechanisms are unclear, and developing the target neuroprotective strategies are urgent. 2,7,2′-trihydroxy-4,4′7′-trimethoxy-1,1′-biphenanthrene (TTB) is a natural product isolated from Cremastra appendiculata (D. Don) Makino and Liparis nervosa (Thunb.) Lindl. TTB has demonstrated potent cytotoxic activity against stomach (HGC-27) and colon (HT-29) cancer cell lines. However, none of the studies have addressed the effects of TTB in NHIE. In the present study, an oxygen-glucose deprivation/reoxygenation (OGD/R)-induced astrocyte injury model was established to investigate the effect of TTB and its potential mechanisms. Our results showed that TTB alleviated the OGD/R-induced reactive oxygen species increase and the intracellular antioxidant capacity of superoxide dismutase activity decrease. Moreover, TTB potentially prolonged the activation state of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway and maintained the protection against oxidative stress in OGD/R-induced astrocytes by inducing the nuclear translocation and up-regulation of Nrf2 along with the enhanced expression of the downstream target gene HO-1. Furthermore, TTB treatment diminished the accumulation of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) expression induced by OGD/R. We also found TTB-treated astrocytes reversed the inhibition of OGD/R on neurite growth of neurons by the astrocyte-neuron coculture system. In conclusion, TTB inhibited the OGD/R-induced astrocyte oxidative stress at least partially through the inhibition of HIF-1α and VEGF via the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Liang Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhichen Zhao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Qimeng Yin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Xiaolu Zhang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
12
|
Wang H, Wei W, Lan X, Liu N, Li Y, Ma H, Sun T, Peng X, Zhuang C, Yu J. Neuroprotective Effect of Swertiamain on Cerebral Ischemia/Reperfusion Injury by Inducing the Nrf2 Protective Pathway. ACS Chem Neurosci 2019; 10:2276-2286. [PMID: 30753053 DOI: 10.1021/acschemneuro.8b00605] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Oxidative stress plays a vital role in the development of cerebral ischemic/reperfusion (I/R). Targeting oxidative stress is proposed to be an effective strategy to treat cerebral I/R injury. Gentiana macrophylla Pall is reported to have a potential protective effect against stroke. Swertiamarin (Swe), an active secoiridoid glycoside compound isolated from Gentiana macrophylla Pall, has been reported to possess antioxidative potential. This study is to explore whether Swe could prevent brain from I/R injury, and the related mechanisms of oxidative stress are also elucidated using mice middle cerebral artery occlusion (MCAO) model and primary hippocampal neurons oxygen-glucose deprivation/reperfusion (OGD/R) model. Swe (25, 100, or 400 mg/kg) was pretreated intraperitoneally for 7 days until establishment of the MCAO model, while hippocampal neurons were maintained in Swe (0.1, 1, or 10 μM) in the entire process of reoxygenation. The results indicated that Swe pretreatment markedly decreased infarct volume, apoptotic neurons, and oxidative damage and promoted neurologic recovery in vivo. It also decreased reactive oxygen species (ROS) and increased cell viability in vitro. Western blot analyses and immunofluorescence staining demonstrated that Swe pretreatment promoted Nrf2 nuclear translocation from Keap1-Nrf2 complex and enhanced the expressions of NAD(P)H: quinone oxidoreductase-1 (NQO1) and heme oxygenase-1 (HO-1) both in vivo and in vitro, while the expressions could be reversed by a Nrf2 inhibitor. The binding mode of Keap1 with Swe was also proposed by covalent molecular docking. Collectively, Swe could be considered as a promising protective agent against cerebral I/R injury through suppressing oxidative stress by activation of the Nrf2 protective pathway.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Wei Wei
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Xiaobing Lan
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Ning Liu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yuxiang Li
- College of Nursing, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Hanxiang Ma
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Xiaodong Peng
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Chunlin Zhuang
- Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, P.R. China
| | - Jianqiang Yu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
13
|
Catalpol Inhibits Ischemia-Induced Premyelinating Oligodendrocyte Damage through Regulation of Intercellular Calcium Homeostasis via Na⁺/Ca 2+ Exchanger 3. Int J Mol Sci 2018; 19:ijms19071925. [PMID: 29966349 PMCID: PMC6073132 DOI: 10.3390/ijms19071925] [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: 05/12/2018] [Revised: 06/15/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
The heightened vulnerability of premyelinating oligodendrocytes (PreOLs) in response to hypoxia⁻ischemia may contribute to perinatal white matter injury and subsequent neurobehavioral dysfunction. Intracellular Ca2+ overload is considered a crucial mechanism predisposing PreOLs to ischemic injury. We previously reported that catalpol, an iridoid glycoside extracted from Rehmannia root, inhibits intracellular Ca2+ overload of PreOLs in an in vitro ischemia model. However, the exact underlying mechanisms remain elusive. In the present study, we aimed to investigate the protective effects of catalpol on PreOLs and to explore the underlying mechanisms involved in the modulation of intracellular Ca2+ homeostasis. Postnatal day 2 (P2) Sprague-Dawley (SD) rats subjected to bilateral common carotid artery ligation followed by exposure to 8% oxygen for 10 min were used as a rat model of neonatal hypoxia⁻ischemia. We found that catalpol significantly improved behavioral functions and prevented PreOL loss and myelination deficit after hypoxia⁻ischemia. Our in vitro studies also confirmed the direct effects of catalpol on oxygen-glucose deprivation (OGD)-induced cell death and arrested maturation of PreOLs. Moreover, we demonstrated that catalpol significantly inhibited intracellular Ca2+ overload and promoted the expression of Na⁺/Ca2+ exchanger 3 (NCX3). Finally, we found that catalpol significantly reduced mitochondrial damage and subsequent extracellular signal-regulated kinase 1/2 (ERK1/2) and poly-ADP-ribose polymerase-1 (PARP-1) activation. Treatment with NCX3-preferring inhibitor 2-[2-[4-(4-nitrobenzyloxy)phenyl]ethyl]isothiourea (KB-R7943) significantly reversed the protective effects of catalpol on PreOLs under OGD. Overall, our data suggest that catalpol protects PreOLs from ischemic injury through regulation of intercellular Ca2+ homeostasis via upregulation of NCX3 activity.
Collapse
|
14
|
Bombesin attenuated ischemia-induced spatial cognitive and synaptic plasticity impairment associated with oxidative damage. Biomed Pharmacother 2018; 103:87-93. [PMID: 29635132 DOI: 10.1016/j.biopha.2018.03.155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 12/23/2022] Open
Abstract
The dysfunction of spatial cognition is a character to various neurological disorders and therapeutic strategy. However, it is limited to known risk factors clinically so far. Gastrin releasing peptide (GRP) signaling is a neuropeptide system mediating emotional memory events. However, the effects of GRP agonist on spatial cognition and hippocampal synaptic plasticity are rarely investigated, especially in pathologic condition. This study was designed to investigate the long-term effects of GRPR agonist, bombesin, against cognitive impairment induced by chronic cerebral ischemia in rats and its possible mechanisms. Our results revealed that bombesin administration (30 μg/kg/day, for 14 continuous days) significantly protected the cognitive and synaptic plasticity impairments as assessed by the Morris water maze and long-term potentiation tests. The mechanism studies demonstrated that bombesin significantly alleviated the decreased activity of total superoxide dismutase (T-SOD), catalase (CAT) and altered the increased the content of malondialdehyde (MDA). Besides, the decreased expression of synapse plasticity-related proteins, calcium- calmodulin- dependent protein kinase II (CaMKII) and synaptophysin (SYP) in the hippocampus were increased with drug treatment. In conclusion, bombesin could protect the oxidative stress and expression of proteins, which were important for synaptic plasticity and cognitive function impairment induced by chronic cerebral ischemia. Our study is presented to provide novel insights into the effects of bombesin on spatial learning and memory, which should be further explored as a potential drug in disorders involving deficits in cognitive function.
Collapse
|
15
|
Wang S, Zhuang C, Du J, Wu C, You H. The presence of MWCNTs reduces developmental toxicity of PFOS in early life stage of zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 222:201-209. [PMID: 28063710 DOI: 10.1016/j.envpol.2016.12.055] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/13/2016] [Accepted: 12/18/2016] [Indexed: 06/06/2023]
Abstract
Both carbon nanotubes (CNTs) and perfluorooctane sulfonate (PFOS) are used widely. There is considerable concern regarding their ecotoxicity. CNTs might interact with PFOS in water and result in different impacts compared with those after single exposures. To our knowledge, the developmental toxicity of PFOS in the presence of multi-walled carbon nanotubes (MWCNTs) in the early life stage of zebrafish (from 3 h post fertilization (hpf) to 96 hpf) was investigated for the first time in this study. The embryos and larvae were exposed to PFOS (0.2, 0.4, 0.8, and 1.6 mg/L), MWCNTs (50 mg/L), and a mixture of both. Compared with PFOS exposure, the adverse effects induced by PFOS on the hatching rate of zebrafish embryos and the heart rate and body length of zebrafish larvae were reduced in the presence of MWCNTs, and mortality and malformation were also alleviated. In addition, zebrafish larvae exposed to PFOS showed decreased activities of superoxide dismutase, catalase, and glutathione peroxidase, as well as decreased levels of reactive oxygen species and malondialdehyde, in the presence of MWCNTs, indicating that oxidative stress and lipid peroxidation was relieved. Thus, the presence of MWCNTs reduces the developmental toxicity of PFOS in the early life stage of zebrafish.
Collapse
Affiliation(s)
- Shutao Wang
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150028, China.
| | - Changlu Zhuang
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150028, China; Life Science and Environmental Science Research Center, Harbin Institute of Commerce, Harbin 150028, China.
| | - Jia Du
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150028, China.
| | - Chuan Wu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150028, China.
| | - Hong You
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150028, China.
| |
Collapse
|
16
|
Chen Y, Zhang Y, Xu M, Luan J, Piao S, Chi S, Wang H. Catalpol alleviates ovalbumin-induced asthma in mice: Reduced eosinophil infiltration in the lung. Int Immunopharmacol 2016; 43:140-146. [PMID: 27992791 DOI: 10.1016/j.intimp.2016.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/22/2016] [Accepted: 12/08/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Radix Rehmanniae Preparata is a traditional Chinese herbal medicine used to treat asthma, and catalpol is one of the main active ingredients in this herb. In the present study, the effects of catalpol on asthma and the underlying mechanism were explored. METHODS Mice with ovalbumin (OVA)-induced asthma were given 5 or 10mg/kg catalpol from Day 15 to Day 28 (intraperitoneal injection). Histopathologic changes were detected by Hematoxylin and Eosin staining and Periodic Acid Schiff staining. The levels of IgE, interleukin (IL)-4, IL-5 and eotaxin were measured by ELISA. The numbers of lymphocytes, monocytes, basophils and eosinophils in the bronchoalveolar lavage fluid were determined by Wright-Giemsa staining. The expression and distribution of eotaxin and C-C chemokine receptor 3 (CCR3) were detected by immunohistochemistry and immunofluorescence. The expression of interleukin-5 receptor α (IL-5Rα) was detected by Western blot assay. RESULTS Catalpol inhibited OVA-induced inflammation and IgE secretion in the lung. OVA-induced type 2 inflammation was suppressed by catalpol as evidenced by decreased levels of IL-4 and IL-5. Moreover, catalpol inhibited the aberrant eosinophil infiltration in the lungs, and also suppressed OVA-induced elevation of eosinophil chemokine eotaxin and its receptor CCR3. In addition, IL-5Rα expression in the bone marrow cells derived from catalpol-treated asthmatic mice was lower than that from the untreated asthmatic mice. CONCLUSION Our study demonstrated that catalpol attenuated OVA-induced asthma and inhibit the infiltration of inflammatory cells, especially eosinophils, into the lung. This study suggests that catalpol may become a promising drug for the treatment of asthma.
Collapse
Affiliation(s)
- Yanyan Chen
- The Second Department of Paediatrics, the First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, People's Republic of China
| | - Yongzheng Zhang
- The Second Department of Paediatrics, the First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, People's Republic of China
| | - Mingyuan Xu
- Department of Pharmacy, the First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, People's Republic of China
| | - Junqi Luan
- The First Department of Paediatrics, the First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, People's Republic of China
| | - Shengai Piao
- The First Department of Paediatrics, the First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, People's Republic of China
| | - Shuang Chi
- Department of Endemic Disease, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Hai Wang
- The Second Department of Paediatrics, the First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, People's Republic of China.
| |
Collapse
|
17
|
Cai Q, Ma T, Li C, Tian Y, Li H. Catalpol Protects Pre-Myelinating Oligodendrocytes against Ischemia-induced Oxidative Injury through ERK1/2 Signaling Pathway. Int J Biol Sci 2016; 12:1415-1426. [PMID: 27994507 PMCID: PMC5166484 DOI: 10.7150/ijbs.16823] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/06/2016] [Indexed: 11/18/2022] Open
Abstract
The vulnerability of pre-myelinating oligodendrocytes (PreOLs) to ischemic injury plays an important role in the pathogenesis and progression of perinatal white matter injury. Although oxidative stress is thought to be a major pathogenic mechanism predisposing the PreOLs to injury, no effective therapies have been identified to date. The present study aimed to investigate the direct protective effects of catalpol, a potent antioxidant and free radical scavenger, on ischemia-induced oxidative damage in PreOLs and to explore whether the ERK1/2 signaling pathway contributed to the protection provided by catalpol. Primary cultures of PreOLs exposed to oxygen-glucose deprivation (OGD) followed by reperfusion were used as an in vitro model of ischemia. Pretreatment with 0.5 mM catalpol for 1 h prior to OGD treatment significantly reversed ischemia-induced apoptosis in PreOLs and myelination deficits by inhibiting intracellular Ca2+ increase, reducing mitochondrial damage, and ameliorating overproduction of reactive oxygen species (ROS). The expression levels of phosphorylated ERK1/2 (p-ERK1/2) and activated poly-ADP-ribose polymerase-1 (PARP-1) were also markedly decreased by catalpol treatment. Blocking the ERK1/2 signaling pathway with the MEK inhibitor U0126 and catalpol significantly protected PreOLs from ROS-mediated apoptosis under OGD. Taken together, these results suggest that catalpol protects PreOLs against ischemia-induced oxidative injury through ERK1/2 signaling pathway. Catalpol may be a candidate for treating ischemic white matter damage.
Collapse
Affiliation(s)
- Qiyan Cai
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, College of Basic Medicine, The Third Military Medical University, Chongqing, China
| | - Teng Ma
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, College of Basic Medicine, The Third Military Medical University, Chongqing, China
| | - Chengren Li
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, College of Basic Medicine, The Third Military Medical University, Chongqing, China
| | - Yanping Tian
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, College of Basic Medicine, The Third Military Medical University, Chongqing, China
| | - Hongli Li
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, College of Basic Medicine, The Third Military Medical University, Chongqing, China
| |
Collapse
|
18
|
Xu Y, Wang S, Miao Q, Jin K, Lou L, Ye X, Xi Y, Ye J. Protective Role of Hinokitiol Against H 2O 2-Induced Injury in Human Corneal Epithelium. Curr Eye Res 2016; 42:47-53. [PMID: 27269503 DOI: 10.3109/02713683.2016.1151530] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE We recently found that hinokitiol has anti-inflammatory activity in human corneal epithelial (HCE) cells. Herein, we investigated the protective role of hinokitiol against H2O2-induced injury in HCE cells and the mechanisms that underlie its action. METHODS HCE cells were incubated with different concentrations of hinokitiol or dimethylsulfoxide (DMSO), which served as a vehicle control, before H2O2 stimulus. The cell viability was evaluated using a cell counting kit-8 (CCK-8) assay. TUNEL, phosphorylated histone γH2A.X, cleaved caspase-3 expression analyses, and location of cytochrome c were conducted to detect cell injury and apoptosis. Reactive oxygen species (ROS), catalase (CAT), superoxide dismutase (SOD), methane dicarboxylic aldehyde (MDA), and total antioxidative capacity (T-AOC) were used to determine oxidative stress. Bcl-2 and Bax protein expressions were measured by western blotting. RESULTS Hinokitiol significantly improved the cell viability, decreased the apoptosis rate, inhibited DNA damage, and reduced cleaved caspase-3 expression and the leakage of cytochrome c from mimitochondrion to cytoplasm of HCE cells against the oxidative stress induced by H2O2. Generation of ROS and MDA and decreased activity of CAT, SOD, and T-AOC were also ameliorated by hinokitiol administration. Moreover, Bcl-2 expression was down-regulated while Bax was up-regulated by H2O2 stimulus, which were reversed by hinokitiol application. CONCLUSION Hinokitiol protects HCE cells against H2O2-induced injury likely by its antioxidant activity and modulating the Bcl-2 signaling pathway.
Collapse
Affiliation(s)
- Yufeng Xu
- a Department of Ophthalmology , The Second Affiliated Hospital of Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Shengzhan Wang
- a Department of Ophthalmology , The Second Affiliated Hospital of Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Qi Miao
- a Department of Ophthalmology , The Second Affiliated Hospital of Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Kai Jin
- a Department of Ophthalmology , The Second Affiliated Hospital of Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Lixia Lou
- a Department of Ophthalmology , The Second Affiliated Hospital of Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Xin Ye
- a Department of Ophthalmology , The Second Affiliated Hospital of Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Yan Xi
- a Department of Ophthalmology , The Second Affiliated Hospital of Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Juan Ye
- a Department of Ophthalmology , The Second Affiliated Hospital of Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| |
Collapse
|
19
|
Xu C, Ding C, Zhou N, Ruan XM, Guo BX. A polysaccharide from Aloe vera L. var. chinensis (Haw.) Berger prevents damage to human gastric epithelial cells in vitro and to rat gastric mucosa in vivo. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.04.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
20
|
An Analysis of the Combination Frequencies of Constituent Medicinal Herbs in Prescriptions for the Treatment of Stroke in Korean Medicine: Determination of a Group of Candidate Prescriptions for Universal Use. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:2674014. [PMID: 27087820 PMCID: PMC4818814 DOI: 10.1155/2016/2674014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 11/17/2022]
Abstract
In contrast to Western medicine, which typically prescribes one medicine to treat a specific disease, traditional East Asian medicine uses any one of a large number of different prescriptions (mixtures of medicinal herbs), according to the patient's characteristics. Although this can be considered an advantage, the lack of a universal prescription for a specific disease is considered a drawback of traditional East Asian medicine. The establishment of universally applicable prescriptions for specific diseases is therefore required. As a basic first step in this process, this study aimed to select prescriptions used in the treatment of stroke and, through the analysis of medicinal herb combination frequencies, select a high-frequency medicinal herb combination group for further experimental and clinical research. As a result, we selected some candidates of a medicinal herb combination and 13 candidates of a medicinal herb for the treatment of stroke.
Collapse
|
21
|
Hamdi Y, Madfai H, Belhareth R, Mokni M, Masmoudi-Kouki O, Amri M. Prenatal exposure to cigarette smoke enhances oxidative stress in astrocytes of neonatal rat. Toxicol Mech Methods 2016; 26:231-7. [PMID: 26998663 DOI: 10.3109/15376516.2016.1156205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Oxidative stress is involved in the pathogenesis of smoking-related disease. Protection of astrocytes from oxidative insult appears essential to maintain brain function. In this study, we have investigated the effect of gestational cigarette exposure on astrocyte survival. Pregnant female were randomly allocated to the control group or to the cigarette smoke group in which they were placed in an exposure chamber and inhale three cigarettes smoke twice a day for a period of 20 days. The control group was kept in the exposure chamber for the same duration, but without exposure to cigarette smoke. Newborn rats from both groups were weighed 24 h after birth and then cerebral hemispheres were collected for astrocyte culture. Incubation of astrocytes isolated from animals exposed to cigarette smoke with 300 μM H2O2 for 1 h induced a significant decrease of the proportion of surviving cells compared to cells isolated form control animals. We have observed that H2O2-treated astroglial cells derived from cigarette smoke exposure showed more reduced superoxide dismutase and catalase activities than H2O2-treated astroglial cells from control animals. In conclusion, this study indicates that astroglial cells derived from newborn rats exposed in utero to cigarette smoke are more vulnerable to oxidative assault than cultured astrocytes obtained from control animals. These results point out the existence of excitotoxic lesions in newborn exposed in utero to cigarette smoke and suggest that despite their high antioxidative activities, astrocytes cannot survive and protect neurons under massive oxidative stress.
Collapse
Affiliation(s)
- Yosra Hamdi
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Hayfa Madfai
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Rym Belhareth
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Meherzia Mokni
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Olfa Masmoudi-Kouki
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Mohamed Amri
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| |
Collapse
|
22
|
Protective Effect of Diospyros kaki against Glucose-Oxygen-Serum Deprivation-Induced PC12 Cells Injury. Adv Pharmacol Sci 2016; 2016:3073078. [PMID: 26941791 PMCID: PMC4749783 DOI: 10.1155/2016/3073078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 01/10/2016] [Indexed: 11/28/2022] Open
Abstract
Ischemic cerebrovascular disease is one of the most common causes of death in the world. Recent interests have been focused on natural antioxidants and anti-inflammatory agents as potentially useful neuroprotective agents. Diospyros kaki (persimmon) has been shown to exert anti-inflammatory, antioxidant, and antineoplastic effects. However, its effects on ischemic damage have not been evaluated. Here, we used an in vitro model of cerebral ischemia and studied the effects of hydroalcoholic extract of peel (PeHE) and fruit pulp (PuHE) of persimmon on cell viability and markers of oxidative damage mainly intracellular reactive oxygen species (ROS) induced by glucose-oxygen-serum deprivation (GOSD) in PC12 cells. GOSD for 6 h produced significant cell death which was accompanied by increased levels of ROS. Pretreatment with different concentrations of PeHE and PuHE (0–500 μg/mL) for 2 and 24 h markedly restored these changes only at high concentrations. However, no significant differences were seen in the protection against ischemic insult between different extracts and the time of exposure. The experimental results suggest that persimmon protects the PC12 cells from GOSD-induced injury via antioxidant mechanisms. Our findings might raise the possibility of potential therapeutic application of persimmon for managing cerebral ischemic and other neurodegenerative disorders.
Collapse
|
23
|
Xue Q, Liu Y, He R, Yang S, Tong J, Li X, Chen Y, Xu X. Lyophilized Powder of Catalpol and Puerarin Protects Neurovascular Unit from Stroke. Int J Biol Sci 2016; 12:367-80. [PMID: 27019622 PMCID: PMC4807157 DOI: 10.7150/ijbs.14059] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/08/2015] [Indexed: 01/06/2023] Open
Abstract
Hunting for an effective medicine for brain stroke has been a medical task in neuroscience for decades. The present research showed that the lyophilized Powder of Catalpol and Puerarin (C-P) in all the tested doses (65.4 mg/kg, 32.7 mg/kg, 16.4 mg/kg) significantly reduced the neurological deficiency, infarct volume and apoptotic cells in ischemic/reperfusion (I/R) rats. It also promoted astrocyte processes and prolonged neuron axons in infarct area. Further, it decreased MDA, NO, NF-κB/p65, TNF-α, IL-1β and IL-6 and enhanced the EPOR and GAF-43. 65.4 mg/kg and 32.7 mg/kg C-P could up-regulated EPO and VEGF significantly. In vitro, 49 μg/mL and 24.5 μg/mL C-P decreased the leakage of sodium fluorescein and increased the activity of γ-GTP. Additionally, it increased SOD and decreased MDA, NO, and LDH and decreased NF-κB/p65, TNF-α, IL-1β and IL-6 and unregulated EPO, EPOR, VEGF, and GAP-43. Only the dose of 49 μg/mL increased TEER and Claudin-5 and turned the typically damaged morphologies of neurons, astrocytes and endothelium into a favorable trend. These data imply that C-P improved the recovery of neurological deficiency in motor, sense, balance and reflex, and protected the whole NVU by anti-oxidative stress, anti-inflammation and up-regulating some protective factors. This research provides a candidate medicine for brain stroke and, at the same time, a pattern for drug study targeting NVU in vitro.
Collapse
Affiliation(s)
- Qiang Xue
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Yang Liu
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Ran He
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Sheng Yang
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Jie Tong
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Xu Li
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Yi Chen
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Xiaoyu Xu
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China;; 2. Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China;; 3. Institute of Chinese Medicine, Southwest University, Chongqing 400715, China
| |
Collapse
|
24
|
Assessment of C-phycocyanin effect on astrocytes-mediated neuroprotection against oxidative brain injury using 2D and 3D astrocyte tissue model. Sci Rep 2015; 5:14418. [PMID: 26399322 PMCID: PMC4585836 DOI: 10.1038/srep14418] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/28/2015] [Indexed: 01/08/2023] Open
Abstract
Drugs are currently being developed to attenuate oxidative stress as a treatment for brain injuries. C-phycocyanin (C-Pc) is an antioxidant protein of green microalgae known to exert neuroprotective effects against oxidative brain injury. Astrocytes, which compose many portions of the brain, exert various functions to overcome oxidative stress; however, little is known about how C-Pc mediates the antioxidative effects of astrocytes. In this study, we revealed that C-Pc intranasal administration to the middle cerebral artery occlusion (MCAO) rats ensures neuroprotection of ischemic brain by reducing infarct size and improving behavioral deficits. C-Pc also enhanced viability and proliferation but attenuated apoptosis and reactive oxygen species (ROS) of oxidized astrocytes, without cytotoxicity to normal astrocytes and neurons. To elucidate how C-Pc leads astrocytes to enhance neuroprotection and repair of ischemia brain, we firstly developed 3D oxidized astrocyte model. C-Pc had astrocytes upregulate antioxidant enzymes such as SOD and catalase and neurotrophic factors BDNF and NGF, while alleviating inflammatory factors IL-6 and IL-1β and glial scar. Additionally, C-Pc improved viability of 3D oxidized neurons. In summary, C-Pc was concluded to activate oxidized astrocytes to protect and repair the ischemic brain with the combinatorial effects of improved antioxidative, neurotrophic, and anti-inflammatory mechanisms.
Collapse
|
25
|
Xu SY, Hu FY, Ren LJ, Chen L, Zhou ZQ, Zhang XJ, Li WP. Dantrolene enhances the protective effect of hypothermia on cerebral cortex neurons. Neural Regen Res 2015; 10:1279-85. [PMID: 26487856 PMCID: PMC4590241 DOI: 10.4103/1673-5374.162761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2015] [Indexed: 01/05/2023] Open
Abstract
Therapeutic hypothermia is the most promising non-pharmacological neuroprotective strategy against ischemic injury. However, shivering is the most common adverse reaction. Many studies have shown that dantrolene is neuroprotective in in vitro and in vivo ischemic injury models. In addition to its neuroprotective effect, dantrolene neutralizes the adverse reaction of hypothermia. Dantrolene may be an effective adjunctive therapy to enhance the neuroprotection of hypothermia in treating ischemic stroke. Cortical neurons isolated from rat fetuses were exposed to 90 minutes of oxygen-glucose deprivation followed by reoxygenation. Neurons were treated with 40 μM dantrolene, hypothermia (at 33°C), or the combination of both for 12 hours. Results revealed that the combination of dantrolene and hypothermia increased neuronal survival and the mitochondrial membrane potential, and reduced intracellular active oxygen cytoplasmic histone-associated DNA fragmentation, and apoptosis. Furthermore, improvements in cell morphology were observed. The combined treatment enhanced these responses compared with either treatment alone. These findings indicate that dantrolene may be used as an effective adjunctive therapy to enhance the neuroprotective effects of hypothermia in ischemic stroke.
Collapse
Affiliation(s)
- Sui-yi Xu
- Postdoctoral Workstation, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Feng-yun Hu
- Department of Neurology, Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li-jie Ren
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Lei Chen
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Zhu-qing Zhou
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Xie-jun Zhang
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| | - Wei-ping Li
- Postdoctoral Workstation, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Department of Brain Center, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University First Affiliated Hospital, Shenzhen, Guangdong Province, China
| |
Collapse
|
26
|
Deciphering the therapeutic mechanisms of Xiao-Ke-An in treatment of type 2 diabetes in mice by a Fangjiomics approach. Acta Pharmacol Sin 2015; 36:699-707. [PMID: 25960133 DOI: 10.1038/aps.2014.138] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/30/2014] [Indexed: 12/23/2022]
Abstract
AIM Xiao-Ke-An (XKA) is a traditional Chinese medicine (TCM) formula for the treatment of type 2 diabetes (T2D), and the effective ingredients and their targets as well as the mechanisms of XKA remain to be elucidated. In this study we investigated the therapeutic mechanisms of XKA in the treatment of T2D in mice using a Fangjiomics approach. METHODS KKAy mice feeding on a high-fat diet were used as models of T2D, and were orally treated with XKA (0.75 or 1.5 g · kg(-1) · d(-1)) for 32 d. Microarray mRNA expression data were obtained from the livers of the mice. Differentially expressed genes (DEGs) were identified by reverse rate analysis and ANOVA analysis. The compounds in XKA were identified by LC-MS analysis or collected from TCM databases. The relationships between the compounds and targets were established by combining the DEGs with information derived from mining literature or herb target databases. Relevant pathways were identified through a Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis using WebGestalt. RESULTS The compound-target-pathway network based on compounds identified by LC-MS analysis (NCA) included 20 constituent compounds, 46 targets and 36 T2D-related pathways, whereas the compound-target-pathway network based on compounds collected from databases (NCD) consisted of 40 compounds, 68 targets and 21 pathways. In the treatment of T2D, XKA might act mainly by improving carbohydrate and lipid metabolism, as well as ameliorating insulin resistance, inflammation and diabetic vascular complications. CONCLUSION The network-based approach reveals complex therapeutic mechanisms of XKA in the treatment of T2D in mice that involve numerous compounds, targets, and signaling pathways.
Collapse
|
27
|
Neuroprotective Effect of Sodium Butyrate against Cerebral Ischemia/Reperfusion Injury in Mice. BIOMED RESEARCH INTERNATIONAL 2015; 2015:395895. [PMID: 26064905 PMCID: PMC4439479 DOI: 10.1155/2015/395895] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/24/2015] [Indexed: 11/28/2022]
Abstract
Sodium butyrate (NaB) is a dietary microbial fermentation product of fiber and serves as an important neuromodulator in the central nervous system. In this study, we further investigated that NaB attenuated cerebral ischemia/reperfusion (I/R) injury in vivo and its possible mechanisms. NaB (5, 10 mg/kg) was administered intragastrically 3 h after the onset of reperfusion in bilateral common carotid artery occlusion (BCCAO) mice. After 24 h of reperfusion, neurological deficits scores were estimated. Morphological examination was performed by electron microscopy and hematoxylin-eosin (H&E) staining. The levels of oxidative stress and inflammatory cytokines were assessed. Apoptotic neurons were measured by TUNEL; apoptosis-related protein caspase-3, Bcl-2, Bax, the phosphorylation Akt (p-Akt), and BDNF were assayed by western blot and immunohistochemistry. The results showed that 10 mg/kg NaB treatment significantly ameliorated neurological deficit and histopathology changes in cerebral I/R injury. Moreover, 10 mg/kg NaB treatment markedly restored the levels of MDA, SOD, IL-1β, TNF-α, and IL-8. 10 mg/kg NaB treatment also remarkably inhibited the apoptosis, decreasing the levels of caspase-3 and Bax and increasing the levels of Bcl-2, p-Akt, and BDNF. This study suggested that NaB exerts neuroprotective effects on cerebral I/R injury by antioxidant, anti-inflammatory, and antiapoptotic properties and BDNF-PI3K/Akt pathway is involved in antiapoptotic effect.
Collapse
|
28
|
An L, Fu J, Zhang T. Reversible effects of vitamins C and E combination on cognitive deficits and oxidative stress in the hippocampus of melamine-exposed rats. Pharmacol Biochem Behav 2015; 132:152-159. [PMID: 25802127 DOI: 10.1016/j.pbb.2015.03.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 12/11/2022]
Abstract
Previous studies showed that the spatial cognitive deficits of rats were induced by chronic melamine exposure, which was associated with the hippocampal oxidative damage. Currently, we examined the antioxidative effect of vitamins C and E combination on cognitive function in melamine-treated rats. Melamine was oral administrated to male adolescent Wistar at a dosage of 300mg/kg/day for 28days. After that, animals received vitamins C and E at a dose of 150 and 200mg/kg, respectively, intraperitoneally for the next 7days. Cognitive behaviors were investigated using the Morris water maze test. The biochemical indexes were detected in the hippocampal homogenate. The treatment with vitamin complex significantly ameliorated cognitive deficits induced by melamine. ROS, MDA, and NO contents were almost back to normal, while SOD, CAT, GSH-Px, and NOS activities were improved as well. The neural apoptosis in the hippocampus were ameliorated by regulating the expression of anti-apoptotic protein (Bcl-2) and caspase-3. Additionally, histological observation showed that vitamin complex effectively alleviated the injuries of hippocampal neurons. These results suggest that the potential therapeutic for oxidative damage induced neuronal apoptosis after treatment of vitamins C and E combination, which is most likely related to the antioxidative effects.
Collapse
Affiliation(s)
- Lei An
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China; Max-Planck Institute for Neurological Research, Cologne 50931, Germany
| | - Jingxuan Fu
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
29
|
Glutathione-Dependent Detoxification Processes in Astrocytes. Neurochem Res 2014; 40:2570-82. [PMID: 25428182 DOI: 10.1007/s11064-014-1481-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 11/10/2014] [Accepted: 11/15/2014] [Indexed: 01/17/2023]
Abstract
Astrocytes have a pivotal role in brain as partners of neurons in homeostatic and metabolic processes. Astrocytes also protect other types of brain cells against the toxicity of reactive oxygen species and are considered as first line of defence against the toxic potential of xenobiotics. A key component in many of the astrocytic detoxification processes is the tripeptide glutathione (GSH) which serves as electron donor in the GSH peroxidase-catalyzed reduction of peroxides. In addition, GSH is substrate in the detoxification of xenobiotics and endogenous compounds by GSH-S-transferases which generate GSH conjugates that are efficiently exported from the cells by multidrug resistance proteins. Moreover, GSH reacts with the reactive endogenous carbonyls methylglyoxal and formaldehyde to intermediates which are substrates of detoxifying enzymes. In this article we will review the current knowledge on the GSH metabolism of astrocytes with a special emphasis on GSH-dependent detoxification processes.
Collapse
|
30
|
MENG CONG, LIU CHUAN, LIU YUANWEI, WU FAN. Oxysophoridine attenuates the injury caused by acute myocardial infarction in rats through anti-oxidative, anti-inflammatory and anti-apoptotic pathways. Mol Med Rep 2014; 11:527-32. [DOI: 10.3892/mmr.2014.2748] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 06/20/2014] [Indexed: 11/06/2022] Open
|
31
|
Wu Y, Liu C, Kuang J, Ge Q, Zhang Y, Wang Z. Overexpression of SmLEA enhances salt and drought tolerance in Escherichia coli and Salvia miltiorrhiza. PROTOPLASMA 2014; 251:1191-9. [PMID: 24595620 DOI: 10.1007/s00709-014-0626-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 02/13/2014] [Indexed: 05/10/2023]
Abstract
Salinity and drought are important abiotic stresses limiting plant growth and development. Late embryogenesis abundant (LEA) proteins are a group of proteins associated with tolerance to water-related stress. We previously cloned an LEA gene, SmLEA, from Salvia miltiorrhiza Bunge. Phylogenetic analysis indicated that SmLEA belongs to Group LEA14, which is involved in the dehydration response. To determine its function in detail, we have now overexpressed SmLEA in Escherichia coli and S. miltiorrhiza. The logarithmic increase in accumulations of SmLEA proteins in E. coli occurred earlier under salinity than under standard conditions. SmLEA-transformed S. miltiorrhiza plants also showed faster root elongation and a lower malondialdehyde concentration than the empty vector control plants did when cultured on MS media supplemented with 60 mM NaCl or 150 mM mannitol. Moreover, SmLEA-overexpressing transgenics experienced a less rapid rate of water loss. Under either salinity or drought, overexpressing plants had greater superoxide dismutase activity and a higher glutathione concentration. These results suggest that SmLEA may be useful in efforts to improve drought and salinity tolerance in S. miltiorrhiza. Our data also provide a good foundation for further studies into the stress resistance mechanism and molecular breeding of this valuable medicinal plant.
Collapse
Affiliation(s)
- Yucui Wu
- Key Laboratory of Medicinal Resource and Natural Pharmaceutical Chemistry of Ministry of Education, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drug in Northwest of China, Shaanxi Normal University, Xi'an, 710062, People's Republic of China
| | | | | | | | | | | |
Collapse
|
32
|
Xu SY, Hu YF, Li WP, Wu YM, Ji Z, Wang SN, Li K, Pan SY. Intermittent hypothermia is neuroprotective in an in vitro model of ischemic stroke. Int J Biol Sci 2014; 10:873-81. [PMID: 25170301 PMCID: PMC4147221 DOI: 10.7150/ijbs.8868] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 07/14/2014] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To investigate whether the intermittent hypothermia (IH) protects neurons against ischemic insult and the potential molecular targets using an in vitro ischemic model of oxygen glucose deprivation (OGD). METHODS Fetal rat cortical neurons isolated from Day E18 rat embryos were subjected to 90-min OGD and hypothermia treatments during reoxygenation before examining the changes in microscopic morphology, cell viability, microtubule- associated protein 2 (MAP-2) release, intracellular pH value and calcium, reactive oxygen species (ROS) generation, mitochondrial membrane potential (△Ψm) and neuronal death using cell counting kit (CCK-8), enzyme-linked immunosorbent assay (ELISA), BCECF AM, Fluo-3 AM, DCFH-DA and dihydroethidium (DHE), JC-1 staining and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), respectively. RESULTS 90-min OGD induced morphologic abnormalities, cell viability decline, MAP-2 release, intracellular acidosis, calcium overload, increased ROS generation, △Ψm decrease and cell death in primary neurons, which was partially inhibited by continuous hypothermia (CH) and intermittent hypothermia (IH). Interestingly, 6-h CH was insufficient to reduce intracellular calcium overload and stabilize mitochondrial membrane potential (△Ψm), while 12-h CH was effective in reversing the above changes. All IH treatments (6×1 h, 4×1.5 h or 3×2 h) effectively attenuated intracellular free calcium overload, inhibited ROS production, stabilized mitochondrial membrane potential (△Ψm) and reduced delayed cell death in OGD-treated cells. However, only IH intervals longer than 1.5 h appeared to be effective in preventing cell viability loss and intracellular pH decline. CONCLUSION Both CH and IH were neuroprotective in an in vitro model of ischemic stroke, and in spite of shorter hypothermia duration, IH could provide a comparable neuroprotection to CH.
Collapse
Affiliation(s)
- Sui-yi Xu
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; ; 2. Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University 1st Affiliated Hospital, Shenzhen 518035, China
| | - Ya-fang Hu
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wei-pin Li
- 2. Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen University 1st Affiliated Hospital, Shenzhen 518035, China
| | - Yong-ming Wu
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhong Ji
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Sheng-nan Wang
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ke Li
- 3. Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Su-yue Pan
- 1. Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
33
|
Shang YH, Tian JF, Hou M, Xu XY. Progress on the protective effect of compounds from natural medicines on cerebral ischemia. Chin J Nat Med 2014; 11:588-95. [PMID: 24345499 DOI: 10.1016/s1875-5364(13)60068-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Indexed: 01/23/2023]
Abstract
The treatment of cerebral ischemic disease by natural medicines has a long history, and has accumulated a rich theoretical knowledge and treatment experience. The objective of this review is to critically evaluate the experimental research situation of the protective effect of the individual compounds from natural medicine on cerebral ischemia in the past ten years, emphasizing the major mechanisms underlying cerebral ischemic pathophysiology. Sixteen representative compounds from natural medicines which are often used to treat stroke are discussed. The results indicate that these components possess a protective effect on cerebral ischemia, and that these components have different mechanisms, including inhibiting excitotoxicity by ginkgolide B, antiapoptosis of breviscapine, influencing astrocytic activation and proliferation of tanshinone IIA, influencing free radicals by ginsenoside Rd, impairing blood-brain barrier disruption by baicalin, and the anti-inflammatory activity of tetramethylpyrazine. Moreover, some components have multiple neuroprotective mechanisms. Therefore, the combination of individual compounds from natural medicines, considering the mechanisms of cerebral ischemia, may be beneficial to patients with cerebral ischemia in the future. This approach will provide a direction for the further application and exploitation of new drug development in the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Yuan-Hong Shang
- College of Pharmaceutical Sciences, Southwest University, Chongqing Engineering Research Center for Pharmacodynamics Evaluation, Chongqing 400716, China; College of Medicine, Panzhihua University, Panzhihua 617000, China
| | - Jin-Feng Tian
- College of Medicine, Panzhihua University, Panzhihua 617000, China
| | - Min Hou
- College of Pharmaceutical Sciences, Southwest University, Chongqing Engineering Research Center for Pharmacodynamics Evaluation, Chongqing 400716, China
| | - Xiao-Yu Xu
- College of Pharmaceutical Sciences, Southwest University, Chongqing Engineering Research Center for Pharmacodynamics Evaluation, Chongqing 400716, China.
| |
Collapse
|
34
|
Liu YR, Li PW, Suo JJ, Sun Y, Zhang BA, Lu H, Zhu HC, Zhang GB. Catalpol provides protective effects against cerebral ischaemia/reperfusion injury in gerbils. J Pharm Pharmacol 2014; 66:1265-70. [PMID: 24720795 DOI: 10.1111/jphp.12261] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 03/02/2014] [Indexed: 12/27/2022]
Abstract
Abstract
Objectives
To investigate the protective effect of catalpol on cerebral ischaemia/reperfusion (CI/R) injury in gerbils and further explore the underlying mechanism.
Methods
A gerbil model of CI/R was prepared by bilateral common carotid occlusion for 10 min followed by 6 h reperfusion. Catalpol (5, 10 or 20 mg/kg per day) was injected intraperitoneally for 3 days before the carotid occlusion. Stroke index was measured during the reperfusion. The contents of endogenous neuropeptides, endothelin-1 (ET-1) and calcitonin gene-related peptide in plasma were evaluated by radioimmunoassay. Superoxide dismutase (SOD) and malondialdehyde (MDA) in brain tissue homogenate were also examined.
Key findings
The results showed that catalpol significantly improved the stroke index compared with CI/R control group (P < 0.05 or P < 0.01). Catalpol significantly increased the activity of SOD at the doses of 10 and 20 mg/kg (P ≤ 0.05), decreased the brain MDA content and the plasma level of ET-1 at the doses of 10 and 20 mg/kg (P ≤ 0.01).
Conclusions
These data suggested that the efficacy of catalpol pretreatment on CI/R injury may be attributed to reduction of free radicals and inhibition of lipid peroxidation and ET-1 production.
Collapse
Affiliation(s)
- Yan-ru Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peng-wei Li
- Pharmacy College, He'nan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jian-jun Suo
- Department of Neurology, The Third People's Hospital of Sanmenxia, Sanmenxia, China
| | - Yan Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo-ai Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong-can Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guo-bin Zhang
- Pharmacy College, He'nan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
35
|
Liu YR, Lei RY, Wang CE, Zhang BA, Lu H, Zhu HC, Zhang GB. Effects of catalpol on ATPase and amino acids in gerbils with cerebral ischemia/reperfusion injury. Neurol Sci 2014; 35:1229-33. [DOI: 10.1007/s10072-014-1687-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 02/13/2014] [Indexed: 10/25/2022]
|
36
|
Sui X, Gao C. Huperzine A ameliorates damage induced by acute myocardial infarction in rats through antioxidant, anti-apoptotic and anti-inflammatory mechanisms. Int J Mol Med 2014; 33:227-233. [PMID: 24190328 DOI: 10.3892/ijmm.2013.1546] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/21/2013] [Indexed: 11/06/2022] Open
Abstract
Huperzine A (HupA), an alkaloid used in traditional Chinese medicine and isolated from Huperzia serrata, has been shown to possess diverse biological activities. The present study was undertaken to evaluate the cardioprotective potential of HupA in myocardial ischemic damage using a rat model of acute myocardial infarction. HupA significantly diminished the infarct size and inhibited the activities of myocardial enzymes, including creatine kinase (CK), the MB isoenzyme of creatine kinase (CK-MB), lactate dehydrogenase (LDH) and cardiac troponin T (cTnT). A significantly reduced activity of malondialdehyde (MDA) and elevated activities of superoxide dismutase (SOD), of the non-enzymatic scavenger enzyme, glutathione (GSH), as well as of glutathione peroxidase (GSH-PX) were found in the HupA-treated groups. Furthermore, decreased protein levels of caspase-3 and Bax, and increased levels of Bcl-2 were observed in the infarcted hearts of the rats treated with various concentrations of HupA. In addition, treatment with HupA markedly inhibited the expression of the nuclear factor-κB (NF-κB) subunit p65, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). These findings suggest that the cardioprotective potential of HupA is associated with its antioxidant, anti-apoptotic and anti-inflammatory properties in acute myocardial infarction in rats.
Collapse
Affiliation(s)
- Xizhong Sui
- Department of Cardiac Surgery, The PLA General Hospital, Medical School of Chinese PLA, Beijing 100098, P.R. China
| | | |
Collapse
|
37
|
An L, Li Z, Zhang T. Reversible effects of vitamins C and E combination on oxidative stress-induced apoptosis in melamine-treated PC12 cells. Free Radic Res 2013; 48:239-50. [PMID: 24182201 DOI: 10.3109/10715762.2013.861598] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Due to its high nitrogen content, melamine was deliberately added to raw milk for increasing the apparent protein content. Previous studies showed that melamine-induced apoptosis and oxidative damage on PC12 cells and rats' hippocampus. Several evidences suggested that vitamin antioxidant reduced oxidative stress and improved organic function. Whether treatments with antioxidant vitamins C or E, otherwise combination of them can attenuate oxidative stress after melamine administration remains to be elucidated. In this study, the reversible effects of vitamin antioxidants was investigated on melamine-induced neurotoxicity in cultured PC12 cells, an in vitro model of neuronal cells. When comparing vitamin C and E, the combination of both statistically increased PC12 cells viability. The results further showed that vitamin complex has effectively reduced the formation of reaction oxygen species, decreased the level of malondialdehyde, and elevated the activities of antioxidative enzymes. Hoechst 33342 staining and flow cytometric analysis of apoptosis showed that vitamin combination treatment effectively prevented PC12 cells from this melamine-induced apoptosis. It revealed the apoptotic nuclear features of the melamine-induced cell death. Additionally, a combination treatment of vitamins effectively inhibited apoptosis via blocking the increased activation of caspase-3. In summary, the vitamin E and C combination treatment could rescue PC12 cells from the injury induced by melamine through the downregulation of oxidative stress and prevention of melamine-induced apoptosis.
Collapse
Affiliation(s)
- L An
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University , Tianjin , P. R. China
| | | | | |
Collapse
|
38
|
Huang C, Cui Y, Ji L, Zhang W, Li R, Ma L, Xing W, Zhou H, Chen B, Yu J, Zhang H. Catalpol decreases peroxynitrite formation and consequently exerts cardioprotective effects against ischemia/reperfusion insult. PHARMACEUTICAL BIOLOGY 2013; 51:463-73. [PMID: 23336403 DOI: 10.3109/13880209.2012.740052] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
CONTEXT Peroxynitrite (ONOO(-)) formation triggers oxidative/nitrative stress and contributes to exacerbated myocardial ischemia/reperfusion (MI/R) injury. Catalpol, an iridoid glycoside, abundantly found in the roots of Rehmannia glutinosa L. that is included in the family Phrymaceae in the order Lamiales, endemic to China, was found to have neuroprotective effects. However, the effect of catalpol on MI/R injury has not been identified. OBJECTIVE This study investigated whether catalpol attenuates oxidative/nitrative stress in acute MI/R. MATERIALS AND METHODS Adult male rats were subjected to 30 min of myocardial ischemia and 3 h of reperfusion and were treated with saline, catalpol (5 mg/kg, i.p., 5 min before reperfusion) or catalpol plus wortmannin (15 µg/kg intraperitoneally injected 15 min before reperfusion). RESULTS Pretreatment with catalpol significantly improved cardiac functions, reduced myocardial infarction, apoptosis and necrosis of cardiomyocytes after MI/R (all p < 0.05). Meanwhile, ONOO(-) formation was markedly reduced after catalpol treatment (3.01 ± 0.22 vs. 4.66 ± 0.53 pmol/mg protein in vehicle, p < 0.05). In addition, catalpol increased Akt and endothelial nitric oxide synthase phosphorylation, nitric oxide (NO) production, anti-oxidant capacity and reduced MI/R-induced inducible nitric oxide synthase expression and superoxide anion (·O(2)(-)) production in I/R hearts. PI3K inhibitor wortmannin not only blocked catalpol-induced Akt activation, but also attenuated all the beneficial effects of catalpol. Suppression of ONOO(-) formation by either catalpol or an ONOO(-) scavenger uric acid (5 mg/kg) reduced myocardial infarct size in MI/R rats. DISCUSSION AND CONCLUSION In conclusion, catalpol affords cardioprotection against MI/R insult by attenuating ONOO(-) formation, which is attributable to increased physiological NO and decreased ·O(2)(-) production.
Collapse
Affiliation(s)
- Chaolian Huang
- Heart Center, First Hospital, Tsinghua University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bi J, Jiang B, Zorn A, Zhao RG, Liu P, An LJ. Catalpol inhibits LPS plus IFN-γ-induced inflammatory response in astrocytes primary cultures. Toxicol In Vitro 2012; 27:543-50. [PMID: 23164921 DOI: 10.1016/j.tiv.2012.09.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 08/14/2012] [Accepted: 09/18/2012] [Indexed: 01/01/2023]
Abstract
A large body of evidence suggests that the inflammatory reaction plays an important role in the pathogenesis of neurodegenerative diseases. Our previous studies described the neuroprotective effects of catalpol in lipopolysaccharide (LPS)-induced inflammatory models, in which catalpol was shown to prevent mesencephalic neuron death and ameliorate cognitive ability animals. To further investigate the protective effect and underlying mechanism of catalpol, astrocytes were pretreated with low (0.1mM) and high dose (0.5mM) catalpol for 1h prior to LPS plus interferon-γ stimulation. Biochemical analyses showed that NO and ROS production and iNOS activity were significantly reduced by catalpol. Data at transcriptional level also demonstrated that catalpol potently attenuated gene expressions involved in inflammation, such as iNOS, COX-2 and TLR4. In addition, our exploration further revealed that the suppressive action of catalpol on inflammation was mediated via inhibiting nuclear factor-κB (NF-κB) activation. Collectively, these results suggest that catalpol can exert inhibitory effects on the inflammatory reaction in astrocytes and that inactivation of NF-κB could be the major determinant for its anti-inflammatory mechanism. Therefore, catalpol may potentially be a highly effective therapeutic agent in treating neurodegenerative diseases associated with inflammation.
Collapse
Affiliation(s)
- Jing Bi
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China
| | | | | | | | | | | |
Collapse
|
40
|
He C, Wang C, Li B, Wu M, Geng H, Chen Y, Zuo Z. Exposure of Sebastiscus marmoratus embryos to pyrene results in neurodevelopmental defects and disturbs related mechanisms. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2012; 116-117:109-115. [PMID: 22487263 DOI: 10.1016/j.aquatox.2012.03.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/13/2012] [Accepted: 03/13/2012] [Indexed: 05/31/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are widespread environmental contaminants, which are known to be carcinogenic and teratogenic. These compounds cause a range of macroscopic malformations, particularly to the craniofacial apparatus and cardiovascular system during vertebrate development. However, little is known concerning microscopic effects, especially on the sensitive early life stages or on the molecular basis of developmental neurotoxicity. Using the rockfish (Sebastiscus marmoratus), we explored the neurodevelopmental defects caused by early-life exposure to environmentally relevant concentrations of pyrene, a 4-ring PAH. The results showed that pyrene substantially disrupted the cranial innervation pattern and caused deficiency of motor nerves. The expression of a protein associated with axon growth, growth associated protein 43, was decreased in the central nervous system after treatment with pyrene. N-methyl-D-aspartate receptor (NMDAR) plays a vital role in a variety of processes, including neuronal development, synaptic plasticity, and neuronal survival and death. Our results showed that the expression of Ca²⁺/calmodulin dependent kinase II and cAMP-response element-binding, which belong to the NMDAR pathway, were increased in a dose-dependent manner after exposure to pyrene. Acetylcholine, an important neurotransmitter which is known to suppress retinal cells neurite outgrowth, was increased by pyrene exposure. Nitric oxide (NO) acts as an activity-dependent retrograde signal that can coordinate axonal targeting and synaptogenesis during development. The level of NO was decreased in a dose-dependent manner following exposure to pyrene. Taken together, the defects in neurodevelopment and the damage to related mechanisms provided the basis for a better understanding of the neurotoxic effects of pyrene.
Collapse
Affiliation(s)
- Chengyong He
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem Research, School of Life Sciences, Xiamen University, Xiamen, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Hamdi Y, Kaddour H, Vaudry D, Douiri S, Bahdoudi S, Leprince J, Castel H, Vaudry H, Amri M, Tonon MC, Masmoudi-Kouki O. The stimulatory effect of the octadecaneuropeptide ODN on astroglial antioxidant enzyme systems is mediated through a GPCR. Front Endocrinol (Lausanne) 2012; 3:138. [PMID: 23181054 PMCID: PMC3502939 DOI: 10.3389/fendo.2012.00138] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Astroglial cells possess an array of cellular defense systems, including superoxide dismutase (SOD) and catalase antioxidant enzymes, to prevent damage caused by oxidative stress on the central nervous system. Astrocytes specifically synthesize and release endozepines, a family of regulatory peptides including the octadecaneuropeptide (ODN). ODN is the ligand of both central-type benzodiazepine receptors (CBR), and an adenylyl cyclase- and phospholipase C-coupled receptor. We have recently shown that ODN is a potent protective agent that prevents hydrogen peroxide (H(2)O(2))-induced inhibition of SOD and catalase activities and stimulation of cell apoptosis in astrocytes. The purpose of the present study was to investigate the type of receptor involved in ODN-induced inhibition of SOD and catalase in cultured rat astrocytes. We found that ODN induced a rapid stimulation of SOD and catalase gene transcription in a concentration-dependent manner. In addition, 0.1 nM ODN blocked H(2)O(2)-evoked reduction of both mRNA levels and activities of SOD and catalase. Furthermore, the inhibitory actions of ODN on the deleterious effects of H(2)O(2) on SOD and catalase were abrogated by the metabotropic ODN receptor antagonist cyclo(1-8)[Dleu(5)]OP, but not by the CBR antagonist flumazenil. Finally, the protective action of ODN against H(2)O(2)-evoked inhibition of endogenous antioxidant systems in astrocytes was protein kinase A (PKA)-dependent, but protein kinase C-independent. Taken together, these data demonstrate for the first time that ODN, acting through its metabotropic receptor coupled to the PKA pathway, prevents oxidative stress-induced alteration of antioxidant enzyme expression and activities. The peptide ODN is thus a potential candidate for the development of specific agonists that would selectively mimic its protective activity.
Collapse
Affiliation(s)
- Yosra Hamdi
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| | - Hadhemi Kaddour
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| | - David Vaudry
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
- International Associated Laboratory Samuel de ChamplainMont-Saint-Aignan, France
- Regional Platform for Cell Imaging of Haute-Normandie, Institute for Medical Research and Innovation, University of RouenMont-Saint-Aignan, France
| | - Salma Douiri
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| | - Seyma Bahdoudi
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| | - Jérôme Leprince
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
- International Associated Laboratory Samuel de ChamplainMont-Saint-Aignan, France
| | - Hélène Castel
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
| | - Hubert Vaudry
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
- International Associated Laboratory Samuel de ChamplainMont-Saint-Aignan, France
- *Correspondence: Mohamed Amri, Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, 2092 Tunis, Tunisia. e-mail: ; Hubert Vaudry, Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, International Associated Laboratory Samuel de Champlain, Regional Platform for Cell Imaging of Haute-Normandie, Institute for Medical Research and Innovation, University of Rouen, 76821 Mont-Saint-Aignan, France. e-mail:
| | - Mohamed Amri
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- *Correspondence: Mohamed Amri, Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El Manar, 2092 Tunis, Tunisia. e-mail: ; Hubert Vaudry, Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, International Associated Laboratory Samuel de Champlain, Regional Platform for Cell Imaging of Haute-Normandie, Institute for Medical Research and Innovation, University of Rouen, 76821 Mont-Saint-Aignan, France. e-mail:
| | - Marie-Christine Tonon
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, University of RouenMont-Saint-Aignan, France
| | - Olfa Masmoudi-Kouki
- Laboratory of Functional Neurophysiology and Pathology, Research Unit UR/11ES09, Department of Biological Sciences, Faculty of Science of Tunis, University Tunis El ManarTunis, Tunisia
| |
Collapse
|
42
|
Wang J, Sun P, Bao Y, Dou B, Song D, Li Y. Vitamin E renders protection to PC12 cells against oxidative damage and apoptosis induced by single-walled carbon nanotubes. Toxicol In Vitro 2011; 26:32-41. [PMID: 22020378 DOI: 10.1016/j.tiv.2011.10.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 08/14/2011] [Accepted: 10/06/2011] [Indexed: 12/26/2022]
Abstract
Single-walled carbon nanotubes (SWCNTs) are potential candidates in many biomedical applications. However, many reports demonstrated its potential toxicity to human and other biological systems. Our study has demonstrated that SWCNTs can induce apoptosis and oxidative damage on PC12 cells, an in vitro model of neuronal cells. In the present study, we for the first time investigated the neuroprotective effects of vitamin E (VE) on SWCNT-induced neurotoxicity in cultured PC12 cells. Vitamin E (0.01-2mM) increased PC12 cells viability and significantly attenuated SWCNTs-induced apoptotic cell death in a time and dose-dependent manner, as demonstrated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, lactate dehydrogenase (LDH) release and morphological observation. The presence of VE inhibited the formation of reactive oxygen species (ROS), decreased the level of lipid peroxide, elevated the level of glutathione (GSH) and activities of superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT). Additionally, VE blocked the reduction in the mitochondrial membrane potential and the activation of caspase-3. VE prevented the down-regulation of Bcl-2 expression and up-regulation of Bax expression induced by SWCNTs in PC12 cells. In summary, VE might protect PC12 cells from the injury induced by SWCNTs through the down-regulation of oxidative stress and prevention of mitochondrial-mediated apoptosis.
Collapse
Affiliation(s)
- Jingyun Wang
- School of Life Science and Biotechnology, State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, PR China.
| | | | | | | | | | | |
Collapse
|
43
|
Traditional chinese medicine for senile dementia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 2012:692621. [PMID: 21808655 PMCID: PMC3144670 DOI: 10.1155/2012/692621] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 05/09/2011] [Indexed: 12/30/2022]
Abstract
Traditional Chinese Medicine (TCM) has a 3000 years' history of human use. A literature survey addressing traditional evidence from human studies was done, with key result that top 10 TCM herb ingredients including Poria cocos, Radix polygalae, Radix glycyrrhizae, Radix angelica sinensis, and Radix rehmanniae were prioritized for highest potential benefit to dementia intervention, related to the highest frequency of use in 236 formulae collected from 29 ancient Pharmacopoeias, ancient formula books, or historical archives on ancient renowned TCM doctors, over the past 10 centuries. Based on the history of use, there was strong clinical support that Radix polygalae is memory improving. Pharmacological investigation also indicated that all the five ingredients mentioned above can elicit memory-improving effects in vivo and in vitro via multiple mechanisms of action, covering estrogen-like, cholinergic, antioxidant, anti-inflammatory, antiapoptotic, neurogenetic, and anti-Aβ activities. Furthermore, 11 active principles were identified, including sinapic acid, tenuifolin, isoliquiritigenin, liquiritigenin, glabridin, ferulic acid, Z-ligustilide, N-methyl-beta-carboline-3-carboxamide, coniferyl ferulate and 11-angeloylsenkyunolide F, and catalpol. It can be concluded that TCM has a potential for complementary and alternative role in treating senile dementia. The scientific evidence is being continuously mined to back up the traditional medical wisdom.
Collapse
|
44
|
Cai QY, Chen XS, Zhan XL, Yao ZX. Protective effects of catalpol on oligodendrocyte death and myelin breakdown in a rat model of chronic cerebral hypoperfusion. Neurosci Lett 2011; 497:22-6. [DOI: 10.1016/j.neulet.2011.04.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 03/30/2011] [Accepted: 04/08/2011] [Indexed: 01/14/2023]
|
45
|
Cao Y, Mao X, Sun C, Zheng P, Gao J, Wang X, Min D, Sun H, Xie N, Cai J. Baicalin attenuates global cerebral ischemia/reperfusion injury in gerbils via anti-oxidative and anti-apoptotic pathways. Brain Res Bull 2011; 85:396-402. [PMID: 21600966 DOI: 10.1016/j.brainresbull.2011.05.002] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Revised: 04/30/2011] [Accepted: 05/04/2011] [Indexed: 12/27/2022]
Abstract
Baicalin is an important medicinal herb purified from the dry roots of Scutellaria baicalensis Georgi. The present study was undertaken to evaluate the neuroprotective effects of baicalin in gerbils subjected to transient global cerebral ischemic-reperfusion injury. Baicalin at doses of 50, 100 and 200mg/kg was intraperitoneally injected into the gerbils immediately after cerebral ischemia. Seven days after reperfusion, hematoxylin and eosin (HE) staining was performed to analyze hippocampal CA1 pyramidal damage histopathologically. In addition, in order to understand the potential protective mechanism of baicalin, we examined anti-oxidative enzymes, such superoxide dismutase (SOD), glutathione peroxidase (GSH-PX), non-enzymatic scavenger glutathione (GSH) and measured the content of malondialdehyde (MDA) in hippocampus. The mRNA and protein expressions of BDNF were determined in ischemic hippocampus by real-time RT-PCR and Western blot, respectively. Evidence for neuronal apoptosis was detected by real-time RT-PCR, Western blot and caspase-3 activity measurement. Histopathological examination showed that the administration of baicalin by the dose of 100 and 200mg/kg significantly attenuated ischemia-induced neuronal cell damage. Reduced level of MDA, obviously elevated activities of SOD and GSH as well as GSH-PX were also found in baicalin-treated groups. Further investigation demonstrated that treatment with baicalin remarkably promoted the expression of BDNF and inhibited the expression of caspase-3 at mRNA and protein levels by real-time RT-PCR and Western blot, respectively. Besides, caspase-3 activity assay also elucidated that the administration of baicalin could significantly suppress caspase-3 in ischemic gerbils hippocampus. Theses findings suggest that baicalin's neuroprotection appears to be associated with its anti-oxidative and anti-apoptotic properties in global cerebral ischemia in the gerbils.
Collapse
Affiliation(s)
- Yonggang Cao
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang J, Sun P, Bao Y, Liu J, An L. Cytotoxicity of single-walled carbon nanotubes on PC12 cells. Toxicol In Vitro 2011; 25:242-50. [DOI: 10.1016/j.tiv.2010.11.010] [Citation(s) in RCA: 157] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 09/28/2010] [Accepted: 11/15/2010] [Indexed: 01/02/2023]
|
47
|
Hong JL, Qin XY, Shu P, Wu G, Wang Q, Qin MJ. Analysis of catalpol derivatives by characteristic neutral losses using liquid chromatography combined with electrospray ionization multistage and time-of-flight mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2010; 24:2680-2686. [PMID: 20740546 DOI: 10.1002/rcm.4676] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
|
48
|
Mu YL, Xie YY, Zhou L, Zhong Y, Liu L, Bai H, Wang YS, Zhang XM. Cardioprotective effect of 'methylamine irisolidone', a new compound, in hypoxia/reoxygenation injury in cultured rat cardiac myocytes. Chem Biodivers 2009; 6:1170-7. [PMID: 19697334 DOI: 10.1002/cbdv.200800314] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
'Methylamine irisolidone' (=5,7-dihydroxy-6-methoxy-3-(4-methoxyphenyl)-8-[(methylamino)methyl]-4H-[1]benzopyran-4-one), a new compound, is a structurally modified kakkalide with good water solubility. In this study, we investigated its effect on hypoxia/reoxygenation (H/R) injury in cultured rat cardiac myocytes. The results showed that methylamine irisolidone could significantly inhibit lactate dehydrogenase (LDH) release, enhance the mitochondrial membrane potential, decrease intracellular calcium (Ca(2+)) associated with the attenuation of reactive oxygen species (ROS) generation, reduce contents of malondialdehyde (MDA), and increase the activity of superoxide dismutase (SOD) after H/R in a dose-dependent manner. The present study demonstrated that methylamine irisolidone can directly protect cardiomyocytes against H/R injury, primarily as a result of reduction of the intracellular Ca(2+) overload coincident with an attenuation of ROS generation and ROS-mediated lipid peroxidation, which may contribute to the preservation of mitochondrion function and antioxidant against H/R injury.
Collapse
Affiliation(s)
- Yan-Ling Mu
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Lu R, Gu Y, Si D, Liu C. Quantitation of catalpol in rat plasma by liquid chromatography/electrospray ionization tandem mass spectrometry and its pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci 2009; 877:3589-94. [PMID: 19744895 DOI: 10.1016/j.jchromb.2009.08.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 07/04/2009] [Accepted: 08/31/2009] [Indexed: 10/20/2022]
Abstract
A sensitive, rapid and specific liquid chromatography/tandem mass spectrometry (LC/MS/MS) assay has been established for the quantitation of catalpol in rat plasma. Plasma samples were treated by precipitating protein with methanol and were chromatographed by a Diamonsil C(18) column (150 mm x 4.6 mm I.D., 5 microm) with the mobile phase consisting of methanol and 10 mM ammonium formate (30:70, v/v). The selected reaction monitoring (SRM) transitions were performed at m/z 380.1-->183.0 for catalpol and m/z 364.3-->167.0 for aucubin (IS) in the positive ion mode with electrospray ionization (ESI) source. Calibration curve was linear over the concentration range of 10-20,000 ng/mL. The mean recovery was 76.5+/-5.2% and the matrix effect ranged from -5.1 to 13.0%. The intra- and inter-day precisions were less than 6.3 and 14.6%, respectively, and the accuracy was within +/-5.6%. Catalpol was stable in possible conditions of storing and handling. The validated method has been successfully applied to determine the plasma concentration of catalpol for a pharmacokinetic study of catalpol after oral administration of 50 mg/kg to rats.
Collapse
Affiliation(s)
- Rong Lu
- Tianjin State Key Laboratory of Pharmacokinetics and Pharmacodynamics, Tianjin Institute of Pharmaceutical Research, 308 An Shan West Road, Tianjin 300193, China
| | | | | | | |
Collapse
|
50
|
Neuroprotective effects of N-stearoyltyrosine on transient global cerebral ischemia in gerbils. Brain Res 2009; 1287:146-56. [DOI: 10.1016/j.brainres.2009.06.070] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2009] [Revised: 06/20/2009] [Accepted: 06/23/2009] [Indexed: 12/29/2022]
|