1
|
Lao XY, Sun YL, Zhao ZJ, Liu J, Ruan XF. Pharmacological effects of betulinic acid and its protective mechanisms on the cardiovascular system. Fitoterapia 2025; 183:106561. [PMID: 40288588 DOI: 10.1016/j.fitote.2025.106561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Betulinic acid (BA), a pentacyclic triterpenoid saponin widely found in plants, has attracted attention for its diverse pharmacological activities. Recent studies highlight its cardioprotective potential, promoting its relevance in cardiovascular research. AIM OF THE REVIEW This review summarizes BA's physicochemical properties, structure-activity relationships, natural sources, and synthesis strategies. It further discusses its pharmacokinetics and toxicity to evaluate its drug development potential, with emphasis on cardioprotective effects and related signaling pathways. METHODS Literature was collected from databases such as PubMed and Web of Science, focusing on studies addressing BA's chemical characteristics, biological activities, pharmacokinetics, and cardiovascular relevance. RESULTS BA exerts cardioprotective effects via multiple signaling pathways, including NRF2, NF-κB, MAPK, and NFAT. These contribute to its antioxidant, anti-inflammatory, anti-apoptotic, and anti-proliferative actions, as well as its enhancement of endothelial function through nitric oxide signaling. BA also reduces lipid accumulation. Combined with favorable physicochemical properties and synthetic accessibility, these findings support BA as a promising multifunctional lead compound in cardiovascular pharmacology. CONCLUSION BA shows strong potential as a cardioprotective natural compound. Although further research is needed to validate its clinical efficacy and safety, its multi-target actions and structural versatility provide a solid basis for development in cardiovascular drug discovery.
Collapse
Affiliation(s)
- Xu Yuan Lao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuan Long Sun
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zhe Jun Zhao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Liu
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Fen Ruan
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Zhang Z, Zhang Y, Peng H, Yu Q, Kang X, Liu Y, Zheng Y, Cheng F, Wang X, Li F. Decoding TGR5: A comprehensive review of its impact on cerebral diseases. Pharmacol Res 2025; 213:107671. [PMID: 39988005 DOI: 10.1016/j.phrs.2025.107671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Currently, unraveling the enigmatic realm of drug targets for cerebral disorders poses a formidable challenge. Takeda G protein-coupled receptor 5 (TGR5), also known as G protein-coupled bile acid receptor 1, is a specific bile acid receptor. Widely distributed across various tissues, TGR5 orchestrates a myriad of biological functions encompassing inflammation, energy metabolism, fatty acid metabolism, immune responses, cellular proliferation, apoptosis, and beyond. Alongside its well-documented implications in liver diseases, obesity, type 2 diabetes, tumors, and cardiovascular diseases, a growing body of evidence accentuates the pivotal role of TGR5 in cerebral diseases. Thus, this comprehensive review aimed to scrutinize the current insights into the pathological mechanisms involving TGR5 in cerebral diseases, while contemplating its potential as a promising therapeutic target for cerebral diseases.
Collapse
Affiliation(s)
- Zehan Zhang
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Yifei Zhang
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Hongye Peng
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Qingqian Yu
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Xiangdong Kang
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Ying Liu
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Yuxiao Zheng
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Fafeng Cheng
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Xueqian Wang
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Feng Li
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| |
Collapse
|
3
|
Kaur K, Narang RK, Singh S. Neuroprotective potential of Betulinic acid against TIO 2NP induced neurotoxicity in zebrafish. Int Immunopharmacol 2024; 138:112604. [PMID: 38968863 DOI: 10.1016/j.intimp.2024.112604] [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: 03/07/2024] [Revised: 06/04/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024]
Abstract
Betulinic acid (BA) is a natural triterpenoid extracted from Bacopa monnieri. BA has been reported to be used as a neuroprotective agent, but their molecular mechanisms are still unknown. Therefore, in this study, we attempted to investigate the precise mechanism of BA for its protective effect against Titanium dioxide nanoparticles (TiO2NP) induced neurotoxicity in zebrafish. Hence, our study observation showed that 10 µg/ml dose of TiO2NP caused a rigorous behavioral deficit in zebrafish. Further, biochemical analysis revealed TiO2NP significantly decreased GSH, and SOD, and increased MDA, AChE, TNF-α, IL-1β, and IL-6 levels, suggesting it triggers oxidative stress and neuroinflammation. However, BA at doses of 2.5,5,10 mg/kg improved behavioral as well as biochemical changes in zebrafish brain. Moreover, BA also significantly raised the levels of DA, NE, 5-HT, and GABA and decreased glutamate levels in TiO2NP-treated zebrafish brain. Our histopathological analysis proved that TiO2NP causes morphological changes in the brain. These changes were expressed by increasing pyknotic neurons, which were dose-dependently reduced by Betulinic acid. Likewise, BA upregulated the levels of NRF-2 and HO-1, which can reduce oxidative stress and neuroinflammation. Thus, our study provides evidence for the molecular mechanism behind the neuroprotective effect of Betulinic acid. Rendering to the findings, we can consider BA as a suitable applicant for the treatment of AD-like symptoms.
Collapse
Affiliation(s)
- Karamjeet Kaur
- Research Scholar, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India, 142001; Research Scholar, I.K. Gujral Punjab Technical University, Jalandhar Punjab, India, 144603
| | - R K Narang
- Nanomedicine Research Centre, Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India, 142001
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India, 142001.
| |
Collapse
|
4
|
Farzan M, Farzan M, Shahrani M, Navabi SP, Vardanjani HR, Amini-Khoei H, Shabani S. Neuroprotective properties of Betulin, Betulinic acid, and Ursolic acid as triterpenoids derivatives: a comprehensive review of mechanistic studies. Nutr Neurosci 2024; 27:223-240. [PMID: 36821092 DOI: 10.1080/1028415x.2023.2180865] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Cognitive deficits are the main outcome of neurological disorders whose occurrence has risen over the past three decades. Although there are some pharmacologic approaches approved for managing neurological disorders, it remains largely ineffective. Hence, exploring novel nature-based nutraceuticals is a pressing need to alleviate the results of neurodegenerative diseases, such as Alzheimer's disease (AD) and other neurodegenerative disorders. Some triterpenoids and their derivates can be considered potential therapeutics against neurological disorders due to their neuroprotective and cognitive-improving effects. Betulin (B), betulinic acid (BA), and ursolic acid (UA) are pentacyclic triterpenoid compounds with a variety of biological activities, including antioxidative, neuroprotective and anti-inflammatory properties. This review focuses on the therapeutic efficacy and probable molecular mechanisms of triterpenoids in damage prevention to neurons and restoring cognition in neurodegenerative diseases. Considering few studies on this concept, the precise mechanisms that mediate the effect of these compounds in neurodegenerative disorders have remained unknown. The findings can provide sufficient information about the advantages of these compounds against neurodegenerative diseases.
Collapse
Affiliation(s)
- Mahan Farzan
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahour Farzan
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mehrdad Shahrani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyedeh Parisa Navabi
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossien Rajabi Vardanjani
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sahreh Shabani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
5
|
Huang Y, Zhu Z, Luo C, Ma C, Zhu L, Kong L, Li R, Wu J, Yuan Z, Yi J. Betulinic acid attenuates cognitive dysfunction, oxidative stress, and inflammation in a model of T-2 toxin-induced brain damage. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:52098-52110. [PMID: 35254615 DOI: 10.1007/s11356-022-19498-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
T-2 toxin is a mycotoxin that has harmful effects on the immune system and cognitive function. Betulinic acid (BA) is a plant-derived pentacyclic lupane-type triterpenoid which possesses a wide spectrum of bioactivities. The study was aimed to explore whether BA has a protective effect on cognitive impairment and oxidative stress caused by T-2 toxin. BA was suspended in 1% soluble starch by continuous intragastric administration for 14 days, then the brain damage in mice was induced by a single intraperitoneal injection of T-2 toxin (4 mg/kg). It was found that BA alleviated the reduction of discrimination index in T-2 toxin-treated mice, and enhanced dopamine (DA), 5-hydroxytryptamine (5-HT), and acetylcholine (ACH) levels of brain neurotransmitter. Meanwhile, BA pretreatment ameliorated oxidative stress through increase of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and glutathione (GSH) levels, and inhibition of the generation of reactive oxygen species (ROS) and malondialdehyde (MDA) in the brain of mice exposed to T-2 toxin. Moreover, BA reduced brain hemorrhage and ecchymosis, improved the mitochondrial morphology, enriched the number of organelles, and inhibited cell apoptosis in brain challenged with T-2 toxin. Furthermore, BA inhibited mRNA expression of pro-inflammatory cytokines such as interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α) as well as enhanced mRNA expression of anti-inflammatory cytokine such as IL-10 in the brain of T-2 toxin-triggered mice. Therefore, BA could improve the cognitive function, enhance the antioxidant capacity, and inhibit the secretion of proinflammatory cytokines in brain, thereby playing a preventive and protective role against brain damage caused by T-2 toxin.
Collapse
Affiliation(s)
- You Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Zihan Zhu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Chenxi Luo
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Chaoyang Ma
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Lijuan Zhu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Li Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Rongfang Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Jing Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
6
|
Musayeva A, Unkrig JC, Zhutdieva MB, Manicam C, Ruan Y, Laspas P, Chronopoulos P, Göbel ML, Pfeiffer N, Brochhausen C, Daiber A, Oelze M, Li H, Xia N, Gericke A. Betulinic Acid Protects from Ischemia-Reperfusion Injury in the Mouse Retina. Cells 2021; 10:cells10092440. [PMID: 34572088 PMCID: PMC8469383 DOI: 10.3390/cells10092440] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/16/2022] Open
Abstract
Ischemia/reperfusion (I/R) events are involved in the pathophysiology of numerous ocular diseases. The purpose of this study was to test the hypothesis that betulinic acid protects from I/R injury in the mouse retina. Ocular ischemia was induced in mice by increasing intraocular pressure (IOP) to 110 mm Hg for 45 min, while the fellow eye served as a control. One group of mice received betulinic acid (50 mg/kg/day p.o. once daily) and the other group received the vehicle solution only. Eight days after the I/R event, the animals were killed and the retinal wholemounts and optic nerve cross-sections were prepared and stained with cresyl blue or toluidine blue, respectively, to count cells in the ganglion cell layer (GCL) of the retina and axons in the optic nerve. Retinal arteriole responses were measured in isolated retinas by video microscopy. The levels of reactive oxygen species (ROS) were assessed in retinal cryosections and redox gene expression was determined in isolated retinas by quantitative PCR. I/R markedly reduced cell number in the GCL and axon number in the optic nerve of the vehicle-treated mice. In contrast, only a negligible reduction in cell and axon number was observed following I/R in the betulinic acid-treated mice. Endothelial function was markedly reduced and ROS levels were increased in retinal arterioles of vehicle-exposed eyes following I/R, whereas betulinic acid partially prevented vascular endothelial dysfunction and ROS formation. Moreover, betulinic acid boosted mRNA expression for the antioxidant enzymes SOD3 and HO-1 following I/R. Our data provide evidence that betulinic acid protects from I/R injury in the mouse retina. Improvement of vascular endothelial function and the reduction in ROS levels appear to contribute to the neuroprotective effect.
Collapse
Affiliation(s)
- Aytan Musayeva
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Johanna C. Unkrig
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Mayagozel B. Zhutdieva
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Yue Ruan
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Panagiotis Laspas
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Panagiotis Chronopoulos
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Marie L. Göbel
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Christoph Brochhausen
- Institute of Pathology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany;
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Andreas Daiber
- Department of Cardiology 1, Laboratory of Molecular Cardiology, University Medical Center, Johannes Gutenberg University Mainz, Building 605, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.D.); (M.O.)
| | - Matthias Oelze
- Department of Cardiology 1, Laboratory of Molecular Cardiology, University Medical Center, Johannes Gutenberg University Mainz, Building 605, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.D.); (M.O.)
| | - Huige Li
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (H.L.); (N.X.)
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (H.L.); (N.X.)
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
- Correspondence: ; Tel.: +49-613-117-8276
| |
Collapse
|
7
|
Lou H, Li H, Zhang S, Lu H, Chen Q. A Review on Preparation of Betulinic Acid and Its Biological Activities. Molecules 2021; 26:5583. [PMID: 34577056 PMCID: PMC8468263 DOI: 10.3390/molecules26185583] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 12/11/2022] Open
Abstract
Betulinic acid, a pentacyclic triterpene, is distributed in a variety of plants, such as birch, eucalyptus and plane trees. It shows a wide spectrum of biological and pharmacological properties, such as anti-inflammatory, antibacterial, antiviral, antidiabetic, antimalarial, anti-HIV and antitumor effects. Among them, the antitumor activity of betulinic acid has been extensively studied. However, obtaining betulinic acid from natural resources can no longer meet the needs of medicine and nutrition, so methods such as chemical synthesis and microbial biotransformation have also been used to prepare betulinic acid. At the same time, with the development of synthetic biology and genetic engineering, and the elucidation of the biosynthetic pathways of terpenoid, the biosynthesis of betulinic acid has also been extensively researched. This article reviews the preparation of betulinic acid and its pharmacological activities, in order to provide a reference for the research and utilization of betulinic acid.
Collapse
Affiliation(s)
| | | | | | | | - Qihe Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; (H.L.); (H.L.); (S.Z.); (H.L.)
| |
Collapse
|
8
|
Zhao Y, Shi X, Wang J, Mang J, Xu Z. Betulinic Acid Ameliorates Cerebral Injury in Middle Cerebral Artery Occlusion Rats through Regulating Autophagy. ACS Chem Neurosci 2021; 12:2829-2837. [PMID: 34296845 DOI: 10.1021/acschemneuro.1c00198] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cerebral ischemic stroke (CIS) is an acute cerebrovascular disease that is caused by the sudden rupture of blood vessels inside the brain and the intervention of reperfusion to the brain, resulting in severe cerebral injury. Autophagy has been reported to be involved in the occurrence and progression of CIS. Betulinic acid (BA) is a pentacyclic triterpene acid mainly extracted from birch bark. Studies have shown the neuroprotective effects of BA. Here, the effect and mechanism of BA on ischemia-reperfusion induced cerebral injury was explored using a CIS model in vivo via 1 h middle cerebral artery occlusion (MCAO) and 24 h reperfusion in rats and in vitro via oxygen-glucose deprivation/reperfusion (OGD/R) of PC12 cells, respectively. We found that BA not only reduced cerebral injury by reducing oxidative stress but also activated the SIRT1/FoxO1 pathway to suppress autophagy and improve cerebral injury in MCAO rats. These results provide a basis for the potential clinical application of BA.
Collapse
Affiliation(s)
- Yuelin Zhao
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Xiaohua Shi
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Jiaoqi Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Jing Mang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Zhongxin Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| |
Collapse
|
9
|
Alcalá‐Vida R, Garcia‐Forn M, Castany‐Pladevall C, Creus‐Muncunill J, Ito Y, Blanco E, Golbano A, Crespí‐Vázquez K, Parry A, Slater G, Samarajiwa S, Peiró S, Di Croce L, Narita M, Pérez‐Navarro E. Neuron type-specific increase in lamin B1 contributes to nuclear dysfunction in Huntington's disease. EMBO Mol Med 2021; 13:e12105. [PMID: 33369245 PMCID: PMC7863407 DOI: 10.15252/emmm.202012105] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 01/08/2023] Open
Abstract
Lamins are crucial proteins for nuclear functionality. Here, we provide new evidence showing that increased lamin B1 levels contribute to the pathophysiology of Huntington's disease (HD), a CAG repeat-associated neurodegenerative disorder. Through fluorescence-activated nuclear suspension imaging, we show that nucleus from striatal medium-sized spiny and CA1 hippocampal neurons display increased lamin B1 levels, in correlation with altered nuclear morphology and nucleocytoplasmic transport disruption. Moreover, ChIP-sequencing analysis shows an alteration of lamin-associated chromatin domains in hippocampal nuclei, accompanied by changes in chromatin accessibility and transcriptional dysregulation. Supporting lamin B1 alterations as a causal role in mutant huntingtin-mediated neurodegeneration, pharmacological normalization of lamin B1 levels in the hippocampus of the R6/1 mouse model of HD by betulinic acid administration restored nuclear homeostasis and prevented motor and cognitive dysfunction. Collectively, our work points increased lamin B1 levels as a new pathogenic mechanism in HD and provides a novel target for its intervention.
Collapse
Affiliation(s)
- Rafael Alcalá‐Vida
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutInstitut de NeurociènciesUniversitat de BarcelonaBarcelonaCatalonia
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCatalonia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Present address:
Laboratory of Cognitive and Adaptive NeuroscienceUMR 7364 (CNRS/Strasbourg University)StrasbourgFrance
| | - Marta Garcia‐Forn
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutInstitut de NeurociènciesUniversitat de BarcelonaBarcelonaCatalonia
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCatalonia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Present address:
Seaver Autism Center for Research and TreatmentIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Carla Castany‐Pladevall
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutInstitut de NeurociènciesUniversitat de BarcelonaBarcelonaCatalonia
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCatalonia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Jordi Creus‐Muncunill
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutInstitut de NeurociènciesUniversitat de BarcelonaBarcelonaCatalonia
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCatalonia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Yoko Ito
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Enrique Blanco
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Arantxa Golbano
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutInstitut de NeurociènciesUniversitat de BarcelonaBarcelonaCatalonia
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCatalonia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Kilian Crespí‐Vázquez
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutInstitut de NeurociènciesUniversitat de BarcelonaBarcelonaCatalonia
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCatalonia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Aled Parry
- Epigenetics ProgrammeThe Babraham InstituteCambridgeUK
| | - Guy Slater
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Shamith Samarajiwa
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
| | - Sandra Peiró
- Vall d'Hebron Institute of OncologyBarcelonaSpain
| | - Luciano Di Croce
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- ICREABarcelonaSpain
| | - Masashi Narita
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Esther Pérez‐Navarro
- Departament de BiomedicinaFacultat de Medicina i Ciències de la SalutInstitut de NeurociènciesUniversitat de BarcelonaBarcelonaCatalonia
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaCatalonia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| |
Collapse
|
10
|
Li W, Sun K, Hu F, Chen L, Zhang X, Wang F, Yan B. Protective effects of natural compounds against oxidative stress in ischemic diseases and cancers via activating the Nrf2 signaling pathway: A mini review. J Biochem Mol Toxicol 2020; 35:e22658. [PMID: 33118292 DOI: 10.1002/jbt.22658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/28/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022]
Abstract
Oxidative stress, an imbalance between reactive oxygen species and antioxidants, has been seen in the pathological states of many disorders such as ischemic diseases and cancers. Many natural compounds (NCs) have long been recognized to ameliorate oxidative stress due to their inherent antioxidant activities. The modulation of oxidative stress by NCs via activating the Nrf2 signaling pathway is summarized in the review. Three NCs, ursolic acid, betulinic acid, and curcumin, and the mechanisms of their cytoprotective effects are investigated in myocardial ischemia, cerebral ischemia, skin cancer, and prostate cancer. To promote the therapeutic performance of NCs with poor water solubility, the formulation approach, such as the nano drug delivery system, is elaborated as well in this review.
Collapse
Affiliation(s)
- Wenji Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Kai Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fang Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China
| | - Longfei Chen
- China National Intellectual Property Administration Patent Re-examination and Invalidation Department Pharmaceutical Division, Beijing, China
| | - Xing Zhang
- Departments of Urology, Yangzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, Jiangsu, China
| | - Fuxing Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China
| | - Bingchun Yan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
11
|
Schirra C, Xia N, Schüffler A, Heck A, Hasselwander S, Förstermann U, Li H. Phosphorylation and activation of endothelial nitric oxide synthase by red fruit (Pandanus conoideus Lam) oil and its fractions. JOURNAL OF ETHNOPHARMACOLOGY 2020; 251:112534. [PMID: 31893533 DOI: 10.1016/j.jep.2019.112534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 10/03/2019] [Accepted: 12/27/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Red fruit (Pandanus conoideus Lam) oil (RFO) is utilized by inhabitants of the Papua Island to treat diseases such as infections, cancer, and cardiovascular disease, but the mechanism of action is unknown. AIM OF THE STUDY We have recently shown that RFO stimulates nitric oxide (NO) production in endothelial cells. The present study was conducted to investigate the molecular mechanism of endothelial NO synthase (eNOS) activation by RFO. MATERIALS AND METHODS NO production by endothelial cells was determined with electron paramagnetic resonance. The vascular function of isolated mouse aorta was examined using a wire myograph system. Phosphorylation of eNOS was studied with Western blot analyses. RESULTS RFO induced concentration-dependent vasodilation in isolated mouse aorta. The vasodilator effect of RFO was lost in endothelium-denuded aorta and in aorta from mice deficient in eNOS. Treatment of human EA.hy 926 endothelial cells with RFO led to an enhancement of eNOS phosphorylation at serine 1177 and NO production. The RFO-induced eNOS phosphorylation and NO production were reduced by inhibitors of Akt or AMPK, but not by an inhibitor of CaMKII. The effects of RFO were decreased by pharmacological inhibition of PI3K, indicating an involvement of the PI3K-Akt pathway. Moreover, acetone-soluble fractions and oily fractions of RFO showed higher efficacies than the RFO polar fraction in activating eNOS. CONCLUSIONS RFO contains highly active compounds that enhance NO production through Akt- or AMPK-mediated eNOS phosphorylation. The increase in endothelial NO production is likely to represent one of the molecular mechanisms responsible for the therapeutic effects of RFO.
Collapse
Affiliation(s)
- Christian Schirra
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anja Schüffler
- Institute for Biotechnology and Drug Research (IBWF gGmbH), Kaiserslautern, Germany
| | - Astrid Heck
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Solveig Hasselwander
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ulrich Förstermann
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany.
| |
Collapse
|
12
|
Yi Guan Jian, a Traditional Chinese Herbal Medicine, Alleviates Carbon Tetrachloride-Induced Liver Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9824728. [PMID: 30891081 PMCID: PMC6390237 DOI: 10.1155/2019/9824728] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/28/2018] [Accepted: 11/18/2018] [Indexed: 12/13/2022]
Abstract
Objective To study the protective effect of Yi Guan Jian (YGJ) on liver injury induced by carbon tetrachloride (CCl4) in mice. Methods 50 ICR mice were randomly divided into 5 groups: control group, carbon tetrachloride (CCl4) group, CCl4 + silymarin Group (200 mg/kg), carbon CCl4 + YGJ Group (11.5, 23 g/kg). Except the mice in the control group and the CCl4 group that were given the same volume of distilled water, the mice in other groups were given the drugs for seven days. 2 hours after the last administration, except the mice in the control group, the mice in other groups had intraperitoneal injection of 0.1% CCl4 vegetable oil solution (10 ml/kg). Mice in control group had intraperitoneal injection of the same volume of vegetable oil. After 18h, the blood and liver were collected. The liver of mice was stained with HE staining, the levels of alanine transaminase (ALT) and glutamic pyruvic transaminas (AST) in serum were detected, malondialdehyde (MDA), superoxide dismutase (SOD), interleukin (IL-6, Il-1β), and tumor necrosis factor (TNF-α) in serum and liver were detected, and the Western blot was used to detect the levels of MAPK/NF-κB pathway. Results YGJ significantly decreased the levels of ALT and AST. The contents of MDA, IL-6, Il-1β, and TNF-α in serum and liver were significantly decreased and the contents of SOD in serum and liver were significantly decreased by YGJ, which significantly improved the pathological changes of liver tissue in mice. The levels of MAPK/NF-κB pathway were significantly decreased by YGJ. Conclusion YGJ has protective effect on CCl4 induced liver injury in mice, which may be related to the MAPK/NF-κB signaling pathway.
Collapse
|
13
|
Cheng Z, Yao W, Zheng J, Ding W, Wang Y, Zhang T, Zhu L, Zhou F. A derivative of betulinic acid protects human Retinal Pigment Epithelial (RPE) cells from cobalt chloride-induced acute hypoxic stress. Exp Eye Res 2018; 180:92-101. [PMID: 30578788 DOI: 10.1016/j.exer.2018.12.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 02/05/2023]
Abstract
The Retinal Pigment Epithelium (RPE) is a monolayer of cells located above the choroid. It mediates human visual cycle and nourishes photoreceptors. Hypoxia-induced oxidative stress to RPE is a vital cause of retinal degeneration such as the Age-related Macular Degeneration. Most of these retinal diseases are irreversible with no efficient treatment, therefore protecting RPE cells from hypoxia stress is an important way to prevent or slow down the progression of retinal degeneration. Betulinic acid (BA) and betulin (BE) are pentacyclic triterpenoids with anti-oxidative property, but little is known about their effect on RPE cells. We investigated the protective effect of BA, BE and their derivatives against cobalt chloride-induced hypoxia stress in RPE cells. Human ARPE-19 cells were exposed to BA, BE and their eighteen derivatives (named as H3H20) that we customized through replacing moieties at C3 and C28 positions. We found that cobalt chloride reduced cell viability, increased Reactive Oxygen Species (ROS) production as well as induced apoptosis and necrosis in ARPE-19 cells. Interestingly, the pretreatment of 3-O-acetyl-glycyl- 28-O-glycyl-betulinic acid effectively protected cells from acute hypoxia stress induced by cobalt chloride. Our immunoblotting results suggested that this derivative attenuated the cobalt chloride-induced activation of Akt, Erk and JNK pathways. All findings were further validated in human primary RPE cells. In summary, this BA derivate has protective effect against the acute hypoxic stress in human RPE cells and may be developed into a candidate agent effective in the prevention of prevalent retinal diseases.
Collapse
Affiliation(s)
- Zhengqi Cheng
- School of Pharmacy, The University of Sydney, NSW, 2006, Australia
| | - Wenjuan Yao
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jian Zheng
- Center for Bioactive Products, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, 150040, China
| | - Weimin Ding
- School of Chemical and Environmental Engineering, Harbin University of Science and Technology, Harbin, 150080, Heilongjiang, China
| | - Yang Wang
- Center for Bioactive Products, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, 150040, China
| | - Ting Zhang
- Save Sight Institute, The University of Sydney, Sydney, NSW, 2000, Australia; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Zhu
- Save Sight Institute, The University of Sydney, Sydney, NSW, 2000, Australia
| | - Fanfan Zhou
- School of Pharmacy, The University of Sydney, NSW, 2006, Australia.
| |
Collapse
|
14
|
Red fruit (Pandanus conoideus Lam) oil stimulates nitric oxide production and reduces oxidative stress in endothelial cells. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
15
|
Wang D, Chen T, Liu F. Betulinic acid alleviates myocardial hypoxia/reoxygenation injury via inducing Nrf2/HO-1 and inhibiting p38 and JNK pathways. Eur J Pharmacol 2018; 838:53-59. [DOI: 10.1016/j.ejphar.2018.08.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 08/11/2018] [Accepted: 08/15/2018] [Indexed: 02/02/2023]
|
16
|
Sarkaki A, Farbood Y, Badavi M, Ghadiri A, Ghasemi Dehcheshmeh M, Mansouri E, Navabi SP. The protective effect of betulinic acid on microvascular responsivity and protein expression in alzheimer disease induced by cerebral micro-injection of beta-amyloid and streptozotocin. Microcirculation 2018; 25:e12503. [PMID: 30178892 DOI: 10.1111/micc.12503] [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: 03/14/2018] [Revised: 07/19/2018] [Accepted: 08/30/2018] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Alzheimer's disease (AD) is mainly caused by accumulation of β-amyloid (Aβ) in vessels or parenchyma of the brain. Accordingly, natural compounds such as betulinic acid (BA) might improve the AD signs by increase in blood flow and through reduction in amyloid plaques. METHODS Intra-hippocampal injection of BA (0.2 and 0.4 μmol/L /10 μL DMSO /rat) was done at intervals of 180 and 10 min before co-microinjection of 0.1 μmol/L Aβ dissolved in PBS (5 μL/rat, hippocampi) and 1.5 mg/kg Streptozotocin dissolved in aCSF (10 μL/rat, lateral ventricles). Cerebro-vascular responsivity tested by Laser Doppler, BBB leakage, Elisa assays of cytokines (TNF-α and IL-10), and Western blot analysis of proteins (BDNF and AchE) in the hippocampus were assessed 1 month after the injections. RESULTS Microvascular reaction and BBB function were significantly impaired in AD rats, which were improved via BA pretreatment. BA could increase BDNF expression and decrease cytokine levels in the hippocampus of AD rats (especially 0.1 μmol/L Aβ: 0.4 μmol/L BA); however, no significant changes were detected in the blotting of AchE among the groups. CONCLUSIONS Betulinic acid could have a role in AD through protecting microcirculation, alleviating inflammation, and up-regulating BDNF expression which is clearer toward 1:4 molar ratios of Aβ to BA.
Collapse
Affiliation(s)
- Alireza Sarkaki
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Badavi
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ata Ghadiri
- Cellular & Molecular Research Center, Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Esrafil Mansouri
- Cellular & Molecular Research Center, Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyedeh Parisa Navabi
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
17
|
Pharmacologic activation of cholinergic alpha7 nicotinic receptors mitigates depressive-like behavior in a mouse model of chronic stress. J Neuroinflammation 2017; 14:234. [PMID: 29197398 PMCID: PMC5712092 DOI: 10.1186/s12974-017-1007-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/20/2017] [Indexed: 02/07/2023] Open
Abstract
Background It has been shown that chronic stress-induced depression is associated with exaggerated inflammatory response in the brain. Alpha7 nicotinic acetylcholine receptors (α7nAChRs) regulate the cholinergic anti-inflammatory pathway, but the role of cholinergic signaling and α7nAChR in chronic stress has not yet been examined. Methods In this study, we used a well-documented model of depression in which mice were exposed to 6 h of restraint stress for 21 consecutive days. Components of cholinergic signaling and TLR4 signaling were analyzed in the hippocampus. The main targets of neuroinflammation and neuronal damage were also evaluated after a series of tests for depression-like behavior. Results Chronic restraint stress (CRS) induced alterations in components of central cholinergic signaling in hippocampus, including increases in choline acetyltransferase protein expression and decreases in nuclear STAT3 signaling. CRS also increased TLR4 signaling activity, interleukin-1β, and tumor necrosis factor-α expression, microglial activation, and neuronal morphologic changes. Cholinergic stimulation with the α7nAChR agonist DMXBA significantly alleviated CRS-induced depressive-like behavior, neuroinflammation, and neuronal damage, but these effects were abolished by the selective α7nAChR antagonist α-bungarotoxin. Furthermore, activation of α7nAChRs restored the central cholinergic signaling function, inhibited TLR4-mediated inflammatory signaling and microglial activity, and increased the number of regulatory T cells in the hippocampus. Conclusions These findings provide evidence that α7nAChR activation mitigates CRS-induced neuroinflammation and cell death, suggesting that α7nAChRs could be a new therapeutic target for the prevention and treatment of depression. Electronic supplementary material The online version of this article (10.1186/s12974-017-1007-2) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Down-regulation of NOX4 by betulinic acid protects against cerebral ischemia-reperfusion in mice. Curr Med Sci 2017; 37:744-749. [DOI: 10.1007/s11596-017-1798-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 06/09/2017] [Indexed: 12/19/2022]
|
19
|
Navabi SP, Sarkaki A, Mansouri E, Badavi M, Ghadiri A, Farbood Y. The effects of betulinic acid on neurobehavioral activity, electrophysiology and histological changes in an animal model of the Alzheimer's disease. Behav Brain Res 2017; 337:99-106. [PMID: 28986104 DOI: 10.1016/j.bbr.2017.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/28/2017] [Accepted: 10/02/2017] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is a common disorder characterized by aggregation and conversion of amyloid beta (Aβ) monomers to fibrils. Betulinic acid (BA) strongly accelerated this pathway through circumventing the oligomeric intermediate state. BA at doses of 0.2 and 0.4μM/10μl/rat (intra-hippocampal or i.h injection, vehicle: DMSO) was bilaterally administrated 180 and 10min before co-administration of Aβ (0.1μM/5μl/rat, i.h injection, vehicle: PBS) and Streptozotocin (STZ, 1.5mg/kg/10μl/rat, intracerebroventricular or i.c.v. injection, vehicle: aCSF). The behavioral assessments (spatial and passive avoidance memory, anxiety, locomotion, depression, and motor coordination), electrophysiological evaluations (hippocampal long- term potentiation (LTP)) as well as histological changes were evaluated 30days after injections. The indices of spatial and passive avoidance memory, anxiety/depression and LTP records were significantly impaired in AD rats in comparison with the sham. Pretreatment of BA (0.4μM) showed a more significant effect on memory, anxiety, all LTP parameters, and histological damage compared to a low dose in contrast to the AD group. Overall, BA pretreatment was able to prevent AD-induced neurobehavioral and LTP deficits in rats and the best effect was observed in molar ratio of 1:4 (Aβ to BA).
Collapse
Affiliation(s)
- Seyedeh Parisa Navabi
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esrafil Mansouri
- Cellular & Molecular Research Center, Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Badavi
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ata Ghadiri
- Cellular & Molecular Research Center, Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoob Farbood
- Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
20
|
Protective effect of pilose antler peptide on cerebral ischemia/reperfusion (I/R) injury through Nrf-2/OH-1/NF-κB pathway. Int J Biol Macromol 2017; 102:741-748. [DOI: 10.1016/j.ijbiomac.2017.04.091] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 12/27/2022]
|
21
|
Lewinska A, Bednarz D, Adamczyk-Grochala J, Wnuk M. Phytochemical-induced nucleolar stress results in the inhibition of breast cancer cell proliferation. Redox Biol 2017; 12:469-482. [PMID: 28334682 PMCID: PMC5362140 DOI: 10.1016/j.redox.2017.03.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 02/25/2017] [Accepted: 03/11/2017] [Indexed: 11/19/2022] Open
Abstract
The nucleolus is a stress sensor and compromised nucleolar activity may be considered as an attractive anticancer strategy. In the present study, the effects of three plant-derived natural compounds, i.e., sulforaphane (SFN), ursolic acid (UA) and betulinic acid (BA) on nucleolar state were investigated in breast cancer cell lines of different receptor status, namely MCF-7, MDA-MB-231 and SK-BR-3 cells. Cytostatic action of phytochemicals against breast cancer cells was observed at low micromolar concentration window (5-20µM) and mediated by elevated p21 levels, and cell proliferation of SFN-, UA- and BA-treated normal human mammary epithelial cells (HMEC) was unaffected. Phytochemical-mediated inhibition of cell proliferation was accompanied by increased levels of superoxide and protein carbonylation that lead to disorganization of A- and B-type lamin networks and alterations in the nuclear architecture. Phytochemicals promoted nucleolar stress as judged by the nucleoplasmic translocation of RNA polymerase I-specific transcription initiation factor RRN3/TIF-IA, inhibition of new rRNA synthesis and decrease in number of nucleoli. Phytochemicals also decreased the levels of NOP2, proliferation-associated nucleolar protein p120, and WDR12 required for maturation of 28S and 5.8S ribosomal RNAs and formation of the 60S ribosome, and phosphorylation of S6 ribosomal protein that may result in diminished translation and inhibition of cell proliferation. In summary, three novel ribotoxic stress stimuli were revealed with a potential to be used in nucleolus-focused anticancer therapy.
Collapse
Affiliation(s)
- Anna Lewinska
- Laboratory of Cell Biology, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland.
| | - Diana Bednarz
- Laboratory of Cell Biology, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
| | | | - Maciej Wnuk
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
| |
Collapse
|
22
|
Yamada T, Steinz MM, Kenne E, Lanner JT. Muscle Weakness in Rheumatoid Arthritis: The Role of Ca 2+ and Free Radical Signaling. EBioMedicine 2017; 23:12-19. [PMID: 28781131 PMCID: PMC5605300 DOI: 10.1016/j.ebiom.2017.07.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
In addition to the primary symptoms arising from inflammatory processes in the joints, muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Muscle weakness not only reduces the quality of life for the affected patients, but also dramatically increases the burden on society since patients' work ability decreases. A 25–70% reduction in muscular strength has been observed in pateints with RA when compared with age-matched healthy controls. The reduction in muscle strength is often larger than what could be explained by the reduction in muscle size in patients with RA, which indicates that intracellular (intrinsic) muscle dysfunction plays an important role in the underlying mechanism of muscle weakness associated with RA. In this review, we highlight the present understanding of RA-associated muscle weakness with special focus on how enhanced Ca2 + release from the ryanodine receptor and free radicals (reactive oxygen/nitrogen species) contributes to muscle weakness, and recent developments of novel therapeutic interventions. Muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Intrinsic muscle weakness is important in the underlying mechanisms of muscle weakness associated with rheumatoid arthritis. Enhanced Ca2 + release and peroxynitrite-induced stress contributes to RA-induced muscle weakness.
Collapse
Affiliation(s)
- Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Maarten M Steinz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
23
|
Alhusban A, Kozak A, Pillai B, Ahmed H, Sayed MA, Johnson MH, Ishrat T, Ergul A, Fagan SC. Mechanisms of acute neurovascular protection with AT1 blockade after stroke: Effect of prestroke hypertension. PLoS One 2017; 12:e0178867. [PMID: 28640888 PMCID: PMC5480858 DOI: 10.1371/journal.pone.0178867] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 05/19/2017] [Indexed: 01/13/2023] Open
Abstract
Stroke is a leading cause of adult disability worldwide. Improving stroke outcome requires an orchestrated interplay that involves up regulation of pro-survival pathways and a concomitant suppression of pro-apoptotic mediators. In this investigation, we assessed the involvement of eNOS in the AT1 blocker-mediated protective and pro-recovery effects in animals with hypertension. We also evaluated the effect of acute eNOS inhibition in hypertensive animals. To achieve these goals, spontaneously hypertensive rats (SHR) were implanted with blood pressure transmitters, and randomized to receive either an eNOS inhibitor (L-NIO) or saline one hour before cerebral ischemia induction. After 3 hours of ischemia, animals were further randomized to receive either candesartan or saline at the time of reperfusion and sacrificed either 24 hours or 7 days later. Candesartan induced an early protective effect that was independent of eNOS inhibition (50% improvement in motor function). However, the protective effect of candesartan was associated with about five fold up regulation of BDNF expression and about three fold reduction in ER stress markers, in an eNOS dependent manner. The early benefit of a single dose of candesartan, present at 24 hours after stroke, was diminished at 7 days, perhaps due to a failure to induce an angiogenic response in these hypertensive animals. In conclusion, our findings demonstrate an early prorecovery effect of candesartan at both functional and molecular levels. Candesartan induced prorecovery signaling was mediated through eNOS. This effect was not maintained at 7 days after experimental ischemia.
Collapse
Affiliation(s)
- Ahmed Alhusban
- Program in Clinical and Experimental Therapeutics- Charlie Norwood VA Medical Center and College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
- College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Anna Kozak
- Program in Clinical and Experimental Therapeutics- Charlie Norwood VA Medical Center and College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
| | - Bindu Pillai
- Program in Clinical and Experimental Therapeutics- Charlie Norwood VA Medical Center and College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
| | - Heba Ahmed
- Program in Clinical and Experimental Therapeutics- Charlie Norwood VA Medical Center and College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
| | - Mohammed A. Sayed
- Program in Clinical and Experimental Therapeutics- Charlie Norwood VA Medical Center and College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
| | - Maribeth H. Johnson
- Departments of Biostatistics, Medical College of Georgia, Augusta University, Augusta, Georgia, Unites States of America
| | - Tauheed Ishrat
- Program in Clinical and Experimental Therapeutics- Charlie Norwood VA Medical Center and College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
| | - Adviye Ergul
- Program in Clinical and Experimental Therapeutics- Charlie Norwood VA Medical Center and College of Pharmacy, University of Georgia, Augusta, Georgia, United States of America
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Susan C. Fagan
- Departments of Biostatistics, Medical College of Georgia, Augusta University, Augusta, Georgia, Unites States of America
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| |
Collapse
|
24
|
Estrogen Receptor Signaling and the PI3K/Akt Pathway Are Involved in Betulinic Acid-Induced eNOS Activation. Molecules 2016; 21:molecules21080973. [PMID: 27463705 PMCID: PMC6273205 DOI: 10.3390/molecules21080973] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 11/16/2022] Open
Abstract
Betulinic acid (BA) is a naturally occurring pentacyclic triterpenoid with anti-inflammatory, antiviral and anti-cancer properties. Beneficial cardiovascular effects such as increased nitric oxide (NO) production through enhancement of endothelial NO synthase (eNOS) activity and upregulation of eNOS expression have been demonstrated for this compound. In the present study, immortalized human EA.hy 926 endothelial cells were incubated for up to 1 h with 1–100 µM BA and with the phosphatidylinositol-3-kinase (PI3K) inhibitors LY294002 and wortmannin, or the estrogen receptor (ER) antagonist ICI 182,780. Phosphorylation status of eNOS and total eNOS protein were analyzed by Western blotting using a serine 1177 phosphosite-specific antibody. Bioactive NO production was assessed by determination of cGMP content in rat lung fibroblasts (RFL-6) reporter cells. Short-term incubation of EA.hy 926 cells with BA resulted in eNOS phosphorylation at the serine 1177 residue in a concentration- and time-dependent manner with a half-maximal effective concentration of 0.57 µM. This was associated with an enhanced production of NO. BA-induced eNOS phosphorylation and NO production was completely blocked by pretreatment with ICI 182,780, and was attenuated by pretreatment with the PI3K inhibitors wortmannin and LY294002. These results indicate that fast non-genomic effects of ER with downstream signaling through the PI3K/Akt pathway and consecutive eNOS phosphorylation at serine 1177 are involved in BA-induced eNOS activation.
Collapse
|
25
|
Li Y, Xu XL, Zhao D, Pan LN, Huang CW, Guo LJ, Lu Q, Wang J. TLR3 ligand Poly IC Attenuates Reactive Astrogliosis and Improves Recovery of Rats after Focal Cerebral Ischemia. CNS Neurosci Ther 2016; 21:905-13. [PMID: 26494128 DOI: 10.1111/cns.12469] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/06/2015] [Accepted: 09/13/2015] [Indexed: 01/08/2023] Open
Abstract
AIMS Brain ischemia activates astrocytes in a process known as astrogliosis. Although this process has beneficial effects, excessive astrogliosis can impair neuronal recovery. Polyinosinic-polycytidylic acid (Poly IC) has shown neuroprotection against cerebral ischemia-reperfusion injury, but whether it regulates reactive astrogliosis and glial scar formation is not clear. METHODS We exposed cultured astrocytes to oxygen-glucose deprivation/reoxygenation (OGD/R) and used a rat middle cerebral artery occlusion (MCAO)/reperfusion model to investigate the effects of Poly IC. Astrocyte proliferation and proliferation-related molecules were evaluated by immunostaining and Western blotting. Neurological deficit scores, infarct volumes and neuroplasticity were evaluated in rats after transient MCAO. RESULTS In vitro, Poly IC inhibited astrocyte proliferation, upregulated Toll-like receptor 3 (TLR3) expression, upregulated interferon-β, and downregulated interleukin-6 production. These changes were blocked by a neutralizing antibody against TLR3, suggesting that Poly IC function is TLR3-dependent. Moreover, in the MCAO model, Poly IC attenuated reactive astrogliosis, reduced brain infarction volume, and improved neurological function. In addition, Poly IC prevented MCAO-induced reductions in soma size, dendrite length, and number of dendritic bifurcations in cortical neurons of the infarct penumbra. CONCLUSIONS By ameliorating astrogliosis-related damage, Poly IC is a potential therapeutic agent for attenuating neuronal damage and promoting recovery after brain ischemia.
Collapse
Affiliation(s)
- Yang Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu-Lin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Dan Zhao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin-Na Pan
- Medical Department of Neurology, The Second Hospital of Nanchang, Nanchang, China
| | - Chun-Wei Huang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lian-Jun Guo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Qing Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
Ingberg E, Dock H, Theodorsson E, Theodorsson A, Ström JO. Method parameters' impact on mortality and variability in mouse stroke experiments: a meta-analysis. Sci Rep 2016; 6:21086. [PMID: 26876353 PMCID: PMC4753409 DOI: 10.1038/srep21086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/13/2016] [Indexed: 12/17/2022] Open
Abstract
Although hundreds of promising substances have been tested in clinical trials,
thrombolysis currently remains the only specific pharmacological treatment for
ischemic stroke. Poor quality, e.g. low statistical power, in the preclinical
studies has been suggested to play an important role in these failures. Therefore,
it would be attractive to use animal models optimized to minimize unnecessary
mortality and outcome variability, or at least to be able to power studies more
exactly by predicting variability and mortality given a certain experimental setup.
The possible combinations of methodological parameters are innumerous, and an
experimental comparison of them all is therefore not feasible. As an alternative
approach, we extracted data from 334 experimental mouse stroke articles and, using a
hypothesis-driven meta-analysis, investigated the method parameters’
impact on infarct size variability and mortality. The use of Swiss and C57BL6 mice
as well as permanent occlusion of the middle cerebral artery rendered the lowest
variability of the infarct size while the emboli methods increased variability. The
use of Swiss mice increased mortality. Our study offers guidance for researchers
striving to optimize mouse stroke models.
Collapse
Affiliation(s)
- Edvin Ingberg
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Hua Dock
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Elvar Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Annette Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Division of Neuro and Inflammation Science, Department of Clinical and Experimental Medicine, Linköping University, Department of Neurosurgery, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Sweden
| | - Jakob O Ström
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro, Sweden.,School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
27
|
Lingaraju MC, Pathak NN, Begum J, Balaganur V, Bhat RA, Ram M, Kumar D, Kumar D, Tandan SK. Betulinic acid negates oxidative lung injury in surgical sepsis model. J Surg Res 2015; 193:856-67. [DOI: 10.1016/j.jss.2014.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/29/2014] [Accepted: 09/04/2014] [Indexed: 01/05/2023]
|
28
|
McCann SK, Dusting GJ, Roulston CL. Nox2 knockout delays infarct progression and increases vascular recovery through angiogenesis in mice following ischaemic stroke with reperfusion. PLoS One 2014; 9:e110602. [PMID: 25375101 PMCID: PMC4222846 DOI: 10.1371/journal.pone.0110602] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 09/23/2014] [Indexed: 12/14/2022] Open
Abstract
Evidence suggests the NADPH oxidases contribute to ischaemic stroke injury and Nox2 is the most widely studied subtype in the context of stroke. There is still conjecture however regarding the benefits of inhibiting Nox2 to improve stroke outcome. The current study aimed to examine the temporal effects of genetic Nox2 deletion on neuronal loss after ischaemic stroke using knockout (KO) mice with 6, 24 and 72 hour recovery. Transient cerebral ischaemia was induced via intraluminal filament occlusion and resulted in reduced infarct volumes in Nox2 KO mice at 24 h post-stroke compared to wild-type controls. No protection was evident at either 6 h or 72 h post-stroke, with both genotypes exhibiting similar volumes of damage. Reactive oxygen species were detected using dihydroethidium and were co-localised with neurons and microglia in both genotypes using immunofluorescent double-labelling. The effect of Nox2 deletion on vascular damage and recovery was also examined 24 h and 72 h post-stroke using an antibody against laminin. Blood vessel density was decreased in the ischaemic core of both genotypes 24 h post-stroke and returned to pre-stroke levels only in Nox2 KO mice by 72 h. Overall, these results are the first to show that genetic Nox2 deletion merely delays the progression of neuronal loss after stroke but does not prevent it. Additionally, we show for the first time that Nox2 deletion increases re-vascularisation of the damaged brain by 72 h, which may be important in promoting endogenous brain repair mechanisms that rely on re-vascularisation.
Collapse
Affiliation(s)
- Sarah K. McCann
- Stroke Injury and Repair Team, O'Brien Institute, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Surgery, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Gregory J. Dusting
- Cytoprotection Pharmacology Program, Centre for Eye Research, the Royal Eye and Ear Hospital, Melbourne, Victoria, Australia
- Department of Ophthalmology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Carli L. Roulston
- Stroke Injury and Repair Team, O'Brien Institute, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, St Vincent's Campus, University of Melbourne, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
29
|
Pan LN, Zhu W, Li Y, Xu XL, Guo LJ, Lu Q, Wang J. Astrocytic Toll-like receptor 3 is associated with ischemic preconditioning-induced protection against brain ischemia in rodents. PLoS One 2014; 9:e99526. [PMID: 24914679 PMCID: PMC4051824 DOI: 10.1371/journal.pone.0099526] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 05/15/2014] [Indexed: 11/18/2022] Open
Abstract
Background Cerebral ischemic preconditioning (IPC) protects brain against ischemic injury. Activation of Toll-like receptor 3 (TLR3) signaling can induce neuroprotective mediators, but whether astrocytic TLR3 signaling is involved in IPC-induced ischemic tolerance is not known. Methods IPC was modeled in mice with three brief episodes of bilateral carotid occlusion. In vitro, IPC was modeled in astrocytes by 1-h oxygen-glucose deprivation (OGD). Injury and components of the TLR3 signaling pathway were measured after a subsequent protracted ischemic event. A neutralizing antibody against TLR3 was used to evaluate the role of TLR3 signaling in ischemic tolerance. Results IPC in vivo reduced brain damage from permanent middle cerebral artery occlusion in mice and increased expression of TLR3 in cortical astrocytes. IPC also reduced damage in isolated astrocytes after 12-h OGD. In astrocytes, IPC or 12-h OGD alone increased TLR3 expression, and 12-h OGD alone increased expression of phosphorylated NFκB (pNFκB). However, IPC or 12-h OGD alone did not alter the expression of Toll/interleukin receptor domain-containing adaptor-inducing IFNβ (TRIF) or phosphorylated interferon regulatory factor 3 (pIRF3). Exposure to IPC before OGD increased TRIF and pIRF3 expression but decreased pNFκB expression. Analysis of cytokines showed that 12-h OGD alone increased IFNβ and IL-6 secretion; 12-h OGD preceded by IPC further increased IFNβ secretion but decreased IL-6 secretion. Preconditioning with TLR3 ligand Poly I:C increased pIRF3 expression and protected astrocytes against ischemic injury; however, cells treated with a neutralizing antibody against TLR3 lacked the IPC- and Poly I:C-induced ischemic protection and augmentation of IFNβ. Conclusions The results suggest that IPC-induced ischemic tolerance is mediated by astrocytic TLR3 signaling. This reprogramming of TLR3 signaling by IPC in astrocytes may play an important role in suppression of the post-ischemic inflammatory response and thereby protect against ischemic damage. The mechanism may be via activation of the TLR3/TRIF/IRF3 signaling pathway.
Collapse
Affiliation(s)
- Lin-na Pan
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Medical Department of Neurology, The Second Hospital of Nanchang, Nanchang, Jiangxi, People's Republic of China
| | - Wei Zhu
- Department of Emergency Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Yang Li
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xu-lin Xu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- The Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Hubei Province, Wuhan, Hubei, People's Republic of China
| | - Lian-jun Guo
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- The Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Hubei Province, Wuhan, Hubei, People's Republic of China
| | - Qing Lu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- The Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Hubei Province, Wuhan, Hubei, People's Republic of China
- * E-mail:
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
30
|
Cardioprotective effect of betulinic Acid on myocardial ischemia reperfusion injury in rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:573745. [PMID: 24963326 PMCID: PMC4055472 DOI: 10.1155/2014/573745] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/01/2014] [Indexed: 12/21/2022]
Abstract
Objectives. This study aims to investigate the effect of betulinic acid (BA) on myocardial ischemia reperfusion/injury in an open-chest anesthetized rat model. Methods. The model was induced by 30 minutes left anterior descending occlusion followed by 2 hours reperfusion. There are six groups in our present study: sham operation group, ischemia/reperfusion group, low-dosage BA group, medium-dosage BA group, high-dosage BA group, and fosinopril sodium group. Rats in the latter four groups were administrated with BA (50, 100, and 200 mg/kg, i.g.) or fosinopril sodium (10 mg/kg, i.g.) once a day for 7 days before operation, respectively. Rats in the former two groups were given the same volume of vehicle (0.5% CMC-Na, i.g.). During the operation, cardiac function was continuously monitored. Serum LDH and CK were measured with colorimetric assays. The expression of Bcl-2 and Bax and the apoptosis of cardiomyocytes were investigated with western blot and TUNEL assay, respectively. Results. Pretreatment with BA improved cardiac function and attenuated LDH and CK activities compared with IR group. Further investigation demonstrated that the expression of Bcl-2 and Bax and TUNEL assay was in line with the above results. Conclusion. BA may reduce the release of LDH and CK, prevent cardiomyocytes apoptosis, and eventually alleviate the extent of the myocardial ischemia/reperfusion injury.
Collapse
|
31
|
Wang Y, Jia J, Ao G, Hu L, Liu H, Xiao Y, Du H, Alkayed NJ, Liu CF, Cheng J. Hydrogen sulfide protects blood-brain barrier integrity following cerebral ischemia. J Neurochem 2014; 129:827-38. [DOI: 10.1111/jnc.12695] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 02/13/2014] [Accepted: 02/18/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Yali Wang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience; Soochow University; Suzhou Jiangsu China
- The Second Affiliated Hospital of Soochow University; Suzhou Jiangsu China
| | - Jia Jia
- College of Pharmaceutical Science; Soochow University; Suzhou Jiangsu China
| | - Guizhen Ao
- College of Pharmaceutical Science; Soochow University; Suzhou Jiangsu China
| | - Lifang Hu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience; Soochow University; Suzhou Jiangsu China
| | - Hui Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience; Soochow University; Suzhou Jiangsu China
| | - Yunqi Xiao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience; Soochow University; Suzhou Jiangsu China
| | - Huaping Du
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience; Soochow University; Suzhou Jiangsu China
| | - Nabil J. Alkayed
- Department of Anesthesiology & Peri-Operative Medicine; Oregon Health & Science University; Portland Oregon USA
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience; Soochow University; Suzhou Jiangsu China
- The Second Affiliated Hospital of Soochow University; Suzhou Jiangsu China
| | - Jian Cheng
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience; Soochow University; Suzhou Jiangsu China
- The Second Affiliated Hospital of Soochow University; Suzhou Jiangsu China
| |
Collapse
|
32
|
Kamble SM, Goyal SN, Patil CR. Multifunctional pentacyclic triterpenoids as adjuvants in cancer chemotherapy: a review. RSC Adv 2014. [DOI: 10.1039/c4ra02784a] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The protective adjuvants in chemotherapy.
Collapse
Affiliation(s)
- Sarika M. Kamble
- Drug Discovery Laboratory
- Department of Pharmacology
- R. C. Patel Institute of Pharmaceutical Education and Research
- Shirpur, Dist. Dhule, India
| | - Sameer N. Goyal
- Drug Discovery Laboratory
- Department of Pharmacology
- R. C. Patel Institute of Pharmaceutical Education and Research
- Shirpur, Dist. Dhule, India
| | - Chandragouda R. Patil
- Drug Discovery Laboratory
- Department of Pharmacology
- R. C. Patel Institute of Pharmaceutical Education and Research
- Shirpur, Dist. Dhule, India
| |
Collapse
|
33
|
Najjar S, Pearlman DM, Devinsky O, Najjar A, Zagzag D. Neurovascular unit dysfunction with blood-brain barrier hyperpermeability contributes to major depressive disorder: a review of clinical and experimental evidence. J Neuroinflammation 2013; 10:142. [PMID: 24289502 PMCID: PMC4220803 DOI: 10.1186/1742-2094-10-142] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 11/15/2013] [Indexed: 12/27/2022] Open
Abstract
About one-third of people with major depressive disorder (MDD) fail at least two antidepressant drug trials at 1 year. Together with clinical and experimental evidence indicating that the pathophysiology of MDD is multifactorial, this observation underscores the importance of elucidating mechanisms beyond monoaminergic dysregulation that can contribute to the genesis and persistence of MDD. Oxidative stress and neuroinflammation are mechanistically linked to the presence of neurovascular dysfunction with blood-brain barrier (BBB) hyperpermeability in selected neurological disorders, such as stroke, epilepsy, multiple sclerosis, traumatic brain injury, and Alzheimer’s disease. In contrast to other major psychiatric disorders, MDD is frequently comorbid with such neurological disorders and constitutes an independent risk factor for morbidity and mortality in disorders characterized by vascular endothelial dysfunction (cardiovascular disease and diabetes mellitus). Oxidative stress and neuroinflammation are implicated in the neurobiology of MDD. More recent evidence links neurovascular dysfunction with BBB hyperpermeability to MDD without neurological comorbidity. We review this emerging literature and present a theoretical integration between these abnormalities to those involving oxidative stress and neuroinflammation in MDD. We discuss our hypothesis that alterations in endothelial nitric oxide levels and endothelial nitric oxide synthase uncoupling are central mechanistic links in this regard. Understanding the contribution of neurovascular dysfunction with BBB hyperpermeability to the pathophysiology of MDD may help to identify novel therapeutic and preventative approaches.
Collapse
Affiliation(s)
- Souhel Najjar
- Department of Neurology, Neuroinflammation Research Group, Epilepsy Center Division, NYU School of Medicine, New York, NY 10016, USA.
| | | | | | | | | |
Collapse
|
34
|
Capacity of HSYA to inhibit nitrotyrosine formation induced by focal ischemic brain injury. Nitric Oxide 2013; 35:144-51. [PMID: 24126016 DOI: 10.1016/j.niox.2013.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 09/25/2013] [Accepted: 10/01/2013] [Indexed: 12/21/2022]
Abstract
Peroxynitrite-mediated protein tyrosine nitration represents a crucial pathogenic mechanism of stroke. Hydroxysafflor yellow A (HSYA) is the most important active component of the safflower plant. Here we assess the neuroprotective efficacy of HSYA and investigate the mechanism through anti-nitrative pathway. Rats were subjected to 60-min ischemia followed by reperfusion. HSYA (2.5-10mg/kg) was injected at 1h after ischemia onset. Other groups received HSYA (10mg/kg) treatment at 3-9h after onset. Infarct volume, brain edema, and neurological score were evaluated at 24h after ischemia. Nitrotyrosine and inducible NO synthase (iNOS) expression, as well as NO level (nitrate/nitrite) in ischemic cortex was examined within 24h after ischemia. The ability of HSYA to scavenge peroxynitrite was evaluated in vitro. Infarct volume was significantly decreased by HSYA (P<0.05), with a therapeutic window of 3h after ischemia at dose of 10mg/kg. HSYA treatment also reduced brain edema and improved neurological score (P<0.05). Nitrotyrosine formation was dose- and time-dependently inhibited by HSYA. The time window of HSYA in decreasing protein tyrosine nitration paralleled its action in infarct volume. HSYA also greatly reduced iNOS expression and NO content at 24h after ischemia, suggesting prevention of peroxynitrite generation from iNOS. In vitro, HSYA blocked authentic peroxynitrite-induced tyrosine nitration in bovine serum albumin and primary cortical neurons. Collectively, our results indicated that post-ischemic HSYA treatment attenuates brain ischemic injury which is at least partially due to reducing nitrotyrosine formation, possibly by the combined mechanism of its peroxynitrite scavenging ability and its reduction in iNOS production.
Collapse
|
35
|
McCann SK, Roulston CL. NADPH Oxidase as a Therapeutic Target for Neuroprotection against Ischaemic Stroke: Future Perspectives. Brain Sci 2013; 3:561-98. [PMID: 24961415 PMCID: PMC4061864 DOI: 10.3390/brainsci3020561] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/14/2013] [Accepted: 03/20/2013] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress caused by an excess of reactive oxygen species (ROS) is known to contribute to stroke injury, particularly during reperfusion, and antioxidants targeting this process have resulted in improved outcomes experimentally. Unfortunately these improvements have not been successfully translated to the clinical setting. Targeting the source of oxidative stress may provide a superior therapeutic approach. The NADPH oxidases are a family of enzymes dedicated solely to ROS production and pre-clinical animal studies targeting NADPH oxidases have shown promising results. However there are multiple factors that need to be considered for future drug development: There are several homologues of the catalytic subunit of NADPH oxidase. All have differing physiological roles and may contribute differentially to oxidative damage after stroke. Additionally, the role of ROS in brain repair is largely unexplored, which should be taken into consideration when developing drugs that inhibit specific NADPH oxidases after injury. This article focuses on the current knowledge regarding NADPH oxidase after stroke including in vivo genetic and inhibitor studies. The caution required when interpreting reports of positive outcomes after NADPH oxidase inhibition is also discussed, as effects on long term recovery are yet to be investigated and are likely to affect successful clinical translation.
Collapse
Affiliation(s)
- Sarah K McCann
- Stroke Injury and Repair Team, O'Brien Institute, St Vincent's Hospital, 42 Fitzroy St, Fitzroy, Melbourne 3065, Australia.
| | - Carli L Roulston
- Stroke Injury and Repair Team, O'Brien Institute, St Vincent's Hospital, 42 Fitzroy St, Fitzroy, Melbourne 3065, Australia.
| |
Collapse
|
36
|
Radermacher KA, Wingler K, Langhauser F, Altenhöfer S, Kleikers P, Hermans JJR, Hrabě de Angelis M, Kleinschnitz C, Schmidt HHHW. Neuroprotection after stroke by targeting NOX4 as a source of oxidative stress. Antioxid Redox Signal 2013; 18:1418-27. [PMID: 22937798 PMCID: PMC3603500 DOI: 10.1089/ars.2012.4797] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Stroke, a leading cause of death and disability, poses a substantial burden for patients, relatives, and our healthcare systems. Only one drug is approved for treating stroke, and more than 30 contraindications exclude its use in 90% of all patients. Thus, new treatments are urgently needed. In this review, we discuss oxidative stress as a pathomechanism of poststroke neurodegeneration and the inhibition of its source, type 4 nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX4), as a conceptual breakthrough in stroke therapy. RECENT ADVANCES Among potential sources of reactive oxygen species (ROS), the NOXes stand out as the only enzyme family that is solely dedicated to forming ROS. In rodents, three cerebrovascular NOXes exist: the superoxide-forming NOX1 and 2 and the hydrogen peroxide-forming NOX4. Studies using NOX1 knockout mice gave conflicting results, which overall do not point to a role for this isoform. Several reports find NOX2 to be relevant in stroke, albeit to variable and moderate degrees. In our hands, NOX4 is, by far, the major source of oxidative stress and neurodegeneration on ischemic stroke. CRITICAL ISSUES We critically discuss the tools that have been used to validate the roles of NOX in stroke. We also highlight the relevance of different animal models and the need for advanced quality control in preclinical stroke research. FUTURE DIRECTIONS The development of isoform-specific NOX inhibitors presents a precious tool for further clarifying the role and drugability of NOX homologues. This could pave the avenue for the first clinically effective neuroprotectant applied poststroke, and even beyond this, stroke could provide a proof of principle for antioxidative stress therapy.
Collapse
Affiliation(s)
- Kim A Radermacher
- Department of Pharmacology, Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Betulinic acid regulates generation of neuroinflammatory mediators responsible for tissue destruction in multiple sclerosis in vitro. Acta Pharmacol Sin 2013; 34:424-31. [PMID: 23377550 DOI: 10.1038/aps.2012.181] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIM To investigate the influences of betulinic acid (BA), a triterpenoid isolated from birch bark, on neuroinflammatory mediators involved in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis in vitro. METHODS Encephalitogenic T cells were prepared from draining lymph nodes and spinal cords of Dark Agouti rats 8 to 10 d after immunization with myelin basic protein (MBP) and complete Freund's adjuvant. Macrophages were isolated from the peritoneal cavity of adult untreated rats. Astrocytes were isolated from neonatal rat brains. The cells were cultured and then treated with different agents. IFN-γ, IL-17, iNOS and CXCL12 mRNA levels in the cells were analyzed with RT-PCR. iNOS and CXCL12 protein levels were detected using immunoblot. NO and ROS generation was measured using Griess reaction and flow cytometry, respectively. RESULTS In encephalitogenic T cells stimulated with MBP (10 μg/mL), addition of BA inhibited IL-17 and IFN-γ production in a dose-dependent manner. The estimated IC(50) values for IL-17 and IFN γ were 11.2 and 63.8 μmol/L, respectively. When the macrophages were stimulated with LPS (10 ng/mL), addition of BA (50 μmol/L) significantly increased ROS generation, and suppressed NO generation. The astrocytes were stimulated with ConASn containing numerous inflammatory mediators, which mimicked the inflammatory milieu within CNS; addition of BA (50 μmol/L) significantly increased ROS generation, and blocked ConASn-induced increases in iNOS and CXCL12 mRNA levels, but did not affect iNOS and CXCL12 protein levels. Importantly, in both the macrophages and astrocytes, addition of BA (50 μmol/L) inhibited lipid peroxidation. CONCLUSION Besides inhibiting encephalitogenic T cell cytokines and reducing NO generation, BA induces tissue-damaging ROS generation within CNS.
Collapse
|
38
|
Chen XM, Chen HS, Xu MJ, Shen JG. Targeting reactive nitrogen species: a promising therapeutic strategy for cerebral ischemia-reperfusion injury. Acta Pharmacol Sin 2013; 34:67-77. [PMID: 22842734 DOI: 10.1038/aps.2012.82] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ischemic stroke accounts for nearly 80% of stroke cases. Recanalization with thrombolysis is a currently crucial therapeutic strategy for re-building blood supply, but the thrombolytic therapy often companies with cerebral ischemia-reperfusion injury, which are mediated by free radicals. As an important component of free radicals, reactive nitrogen species (RNS), including nitric oxide (NO) and peroxynitrite (ONOO(-)), play important roles in the process of cerebral ischemia-reperfusion injury. Ischemia-reperfusion results in the production of nitric oxide (NO) and peroxynitrite (ONOO(-)) in ischemic brain, which trigger numerous molecular cascades and lead to disruption of the blood brain barrier and exacerbate brain damage. There are few therapeutic strategies available for saving ischemic brains and preventing the subsequent brain damage. Recent evidence suggests that RNS could be a therapeutic target for the treatment of cerebral ischemia-reperfusion injury. Herein, we reviewed the recent progress regarding the roles of RNS in the process of cerebral ischemic-reperfusion injury and discussed the potentials of drug development that target NO and ONOO(-) to treat ischemic stroke. We conclude that modulation for RNS level could be an important therapeutic strategy for preventing cerebral ischemia-reperfusion injury.
Collapse
|
39
|
Toll-like receptor 3 agonist Poly I:C protects against simulated cerebral ischemia in vitro and in vivo. Acta Pharmacol Sin 2012; 33:1246-53. [PMID: 22983393 DOI: 10.1038/aps.2012.122] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AIM To examine the neuroprotective effects of the Toll-like receptor 3 (TLR3) agonist Poly I:C in acute ischemic models in vitro and in vivo. METHODS Primary astrocyte cultures subjected to oxygen-glucose deprivation (OGD) were used as an in vitro simulated ischemic model. Poly I:C was administrated 2 h before OGD. Cell toxicity was measured using MTT assay and LDH leakage assay. The levels of TNFα, IL-6 and interferon-β (IFNβ) in the media were measured using ELISA. Toll/interleukin receptor domain-containing adaptor-inducing IFNβ (TRIF) protein levels were detected using Western blot analysis. A mouse middle cerebral artery occlusion (MCAO) model was u sed for in vivo study. The animals were administered Poly I:C (0.3 mg/kg, im) 2 h before MCAO, and examined with neurological deficit scoring and TTC staining. The levels of TNFα and IL-6 in ischemic brain were measured using ELISA. RESULTS Pretreatment with Poly I:C (10 and 20 μg/mL) markedly attenuated OGD-induced astrocyte injury, and significantly raised the cell viability and reduced the LDH leakage. Poly I:C significantly upregulated TRIF expression accompanied by increased downstream IFNβ production. Moreover, Poly I:C significantly suppressed the pro-inflammatory cytokines TNFα and IL-6 production. In mice subjected to MCAO, administration of Poly I:C significantly attenuated the neurological deficits, reduced infarction volume, and suppressed the increased levels of TNFα and IL-6 in the ischemic striatum and cortex. CONCLUSION Poly I:C pretreatment exerts neuroprotective and anti-inflammatory effects in the simulated cerebral ischemia models, and the neuroprotection is at least in part due to the activation of the TLR3-TRIF pathway.
Collapse
|
40
|
Qian LB, Fu JY, Cai X, Xia ML. Betulinic acid inhibits superoxide anion-mediated impairment of endothelium-dependent relaxation in rat aortas. Indian J Pharmacol 2012; 44:588-592. [PMID: 23112419 PMCID: PMC3480790 DOI: 10.4103/0253-7613.100382] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 04/18/2012] [Accepted: 07/04/2012] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVES To investigate the protective effect of betulinic acid (BA) on endothelium-dependent relaxation (EDR) in rat aortas exposed to pyrogallol-produced superoxide anion and its underlying mechanism. MATERIALS AND METHODS The thoracic aorta of male Sprague-Dawley rats was isolated to mount in the organ bath system and the effect of BA on acetylcholine (ACh)-induced EDR, nitric oxide (NO) level, reactive oxygen species (ROS) level, nitric oxide synthase (NOS) activity, and superoxide dismutase (SOD) activity of aortic rings exposed to pyrogallol (500 μM) for 15 min were measured. RESULTS BA evoked a concentration-dependent EDR in aortas, and pretreatment with EC(50) (2.0 μM) concentration of BA markedly enhanced ACh-induced EDR of aortas exposed to pyrogallol-produced superoxide anion (E(max) rose from 23.91 ± 5.41% to 42.45 ± 9.99%), which was markedly reversed by both N(w) -nitro-L-arginine methyl ester hydrochloride (L-NAME) and methylene blue, but not by indomethacin. Moreover, BA significantly inhibited the increase of ROS level, as well as the decrease of NO level, the endothelial NOS (eNOS) activity, and the SOD activity in aortas induced by pyrogallol-derived superoxide anion. CONCLUSION These results indicate that BA reduces the impairment of EDR in rat aortas exposed to exogenous superoxide anion, which may closely relate to the reduction of oxidative stress and activation of eNOS-NO pathway.
Collapse
Affiliation(s)
- Ling-Bo Qian
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Yin Fu
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Physiological Function, Medical College of Jiaxing University, Jiaxing, China
| | - Xin Cai
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Physiological Function, Medical College of Jiaxing University, Jiaxing, China
| | - Man-Li Xia
- Institute of Physiological Function, Medical College of Jiaxing University, Jiaxing, China
| |
Collapse
|
41
|
Harding SV, Rideout TC, Jones PJH. Evidence for Using Alpha-Lipoic Acid in Reducing Lipoprotein and Inflammatory Related Atherosclerotic Risk. J Diet Suppl 2012; 9:116-27. [DOI: 10.3109/19390211.2012.683136] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Scott V. Harding
- 1Diabetes and Nutritional Sciences Division, School of Medicine, King's College London,
London, UK
| | - Todd C. Rideout
- 2Department of Exercise and Nutrition Sciences, University at Buffalo,
Buffalo, NY, USA
| | - Peter J. H. Jones
- 3Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba,
Winnipeg, Manitoba, Canada
| |
Collapse
|
42
|
Lee JW, Choi YJ, Kim SI, Lee SY, Kang SS, Kim NH, Kwon YS, Lee HJ, Chun WJ, Kim SS. Betulinic Acid Inhibits LPS-Induced MMP-9 Expression by Suppressing NF-kB Activation in BV2 Microglial Cells. Biomol Ther (Seoul) 2011. [DOI: 10.4062/biomolther.2011.19.4.431] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
43
|
Srinivasan K, Sharma SS. 3-Bromo-7-nitroindazole attenuates brain ischemic injury in diabetic stroke via inhibition of endoplasmic reticulum stress pathway involving CHOP. Life Sci 2011; 90:154-60. [PMID: 22075494 DOI: 10.1016/j.lfs.2011.10.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 10/12/2011] [Accepted: 10/24/2011] [Indexed: 12/19/2022]
Abstract
AIMS The role of nitric oxide (NO) and endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of cerebral ischemic/reperfusion (I/R) injury and diabetes. The aim of the study was to investigate the neuroprotective potential of 3-bromo-7-nitroindazole (3-BNI), a potent and selective neuronal nitric oxide synthase (nNOS) inhibitor against ER stress and focal cerebral I/R injury associated with comorbid type 2 diabetes in-vivo. MAIN METHODS Type 2 diabetes was induced by feeding high-fat diet and streptozotocin (35 mg/kg) treatment in rats. Focal cerebral ischemia was induced by 2h middle cerebral artery occlusion (MCAO) followed by 22 h of reperfusion. Immunohistochemistry and western blotting methods were employed for the detection and expression of ER stress/apoptosis markers [78 kDa glucose regulated protein (GRP78) and CCAAT/enhancer binding protein homologous protein (CHOP)]. TUNEL assay for DNA fragmentation was also performed. KEY FINDINGS The diabetic rats subjected to cerebral I/R had prominent neurological damage and functional deficits compared with sham-operated rats. Massive DNA fragmentation was observed in ischemic penumbral region of diabetic brains. Concomitantly, the enhanced immunoreactivity and expression of ER stress/apoptosis markers were noticed. 3-BNI (30 mg/kg, i.p.) treatment significantly inhibited the cerebral infarct, edema volume and improved functional recovery of neurological deficits. The neuroprotection was further evident by lesser DNA fragmentation with a concomitant reduction of GRP78 and CHOP. SIGNIFICANCE The study demonstrates the neuroprotective potential of 3-BNI in diabetic stroke model which may be partly due to inhibition of ER stress pathway involving CHOP.
Collapse
Affiliation(s)
- Krishnamoorthy Srinivasan
- Molecular Neuropharmacology Laboratory, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Sector 67, S.A.S. Nagar, Punjab-160062, India
| | | |
Collapse
|