1
|
Wang Z, Huang C, Shi Z, Liu H, Han X, Chen Z, Li S, Wang Z, Huang J. Coaxial bioprinting of a three-layer vascular structure exhibiting blood-brain barrier function for neuroprotective drug screening. Colloids Surf B Biointerfaces 2025; 249:114494. [PMID: 39787741 DOI: 10.1016/j.colsurfb.2025.114494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/18/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025]
Abstract
The in vitro blood-brain barrier (BBB) structures can offer advantages for studying cerebrovascular functions and developing neuroprotective drugs. However, currently developed BBB models are overly simplistic and inadequate for replicating the complex three-dimensional architecture of the in vivo BBB. In this study, a method is introduced for fabricating a three-layer vascular structure exhibiting BBB function using a coaxial extrusion bioprinting technique with a two-layer nozzle. Photocurable materials were incorporated into the inner layer of the coaxial nozzle, and photoinitiators from the outer layer diffused into the inner layer. As a result, only the materials in the inner layer at the interface between the inner and outer layers underwent crosslinking upon UV exposure. After removing the uncrosslinked materials, a two-layer vascular structure can be formed. Subsequently, a three-layer structure was established after seeding endothelial cells. The perfusion experiments demonstrated that the vascular structure facilitated the continuous flow of culture medium, thereby providing nutrients and oxygen to the surrounding neural tissue. The drug screening analysis indicated that this vascular structure could possess barrier function, allowing the passage of small molecular drugs while effectively blocking macromolecular drugs. Overall, these results suggest that the three-layer vascular structure exhibits excellent perfusion capacity and barrier function, making it a promising candidate for neuroprotective drug screening.
Collapse
Affiliation(s)
- Zhichao Wang
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Chuanzhen Huang
- School of Mechanical Engineering, Yanshan University, Qinhuangdao 066004, China.
| | - Zhenyu Shi
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, China.
| | - Hanlian Liu
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China.
| | - Xu Han
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Zhuang Chen
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Shuying Li
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Zhen Wang
- School of Mechanical Engineering, Yanshan University, Qinhuangdao 066004, China
| | - Jun Huang
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| |
Collapse
|
2
|
Jiao H, Chen R, Jiang Z, Zhang L, Wang H. miR-22 protect PC12 from ischemia/reperfusion-induced injury by targeting p53 upregulated modulator of apoptosis (PUMA). Bioengineered 2020; 11:209-218. [PMID: 32065044 PMCID: PMC7039629 DOI: 10.1080/21655979.2020.1729321] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs have been implicated as critical regulatory molecules in many cerebrovascular diseases. Recent studies demonstrated miR-22 might provide a potential neuroprotective effect. However, the neuroprotective effect of miR-22 in ischemia/reperfusion (I/R) injury has not been thoroughly elucidated. In this study, the PC12 cells were subjected to 4 h oxygen and glucose deprivation (I) and 24 h reoxygenation (R). The PC12 cells were pre-transfected with miR-22 or anti-miR-22 or siRNA-mediated downregulation of p53-upregulated-modulator-of-apoptosis (PUMA)(PUMA siRNA) or their controls at 24 h prior to exposure to I/R. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot were employed to analyze mRNA and protein expression. PI and Annexin V assays and terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) assay were used to quantify the rate of apoptosis. We found that miR-22 expression was significantly downregulated in the PC12 cells subjected to I/R. Loss of function of miR-22 increased PC12 apoptosis after I/R, and overexpression of miR-22 decreases PC12 apoptosis after I/R. PUMA protein was upregulated in the I/R group as compared with the sham group. The increased PUMA protein expression and apoptosis induced by I/R was reversed by transfection with PUMA siRNA. We concluded that I/R enhanced apoptosis and PUMA expression in PC12 cells via downregulation of miR-22. Enhanced miR-22 expression reversed both PUMA expression and apoptosis induced by I/R in PC12 cells. miR-22/PUMA axis has important implications for their clinical applications.
Collapse
Affiliation(s)
- Hongmei Jiao
- Department of Anesthesia, Linyi Cancer Hospital, Linyi, Shandong, China
| | - Renyi Chen
- Department of Anesthesia, Linyi Cancer Hospital, Linyi, Shandong, China
| | - Ziru Jiang
- External Abdominal Section, Linyi Cancer Hospital, Linyi, Shandong, China
| | - Lin Zhang
- Department of Anesthesia, Linyi Cancer Hospital, Linyi, Shandong, China
| | - Hongwei Wang
- Department of Anesthesia, Linyi Cancer Hospital, Linyi, Shandong, China
| |
Collapse
|
3
|
Ihara H, Kakihana Y, Yamakage A, Kai K, Shibata T, Nishida M, Yamada KI, Uchida K. 2-Oxo-histidine-containing dipeptides are functional oxidation products. J Biol Chem 2018; 294:1279-1289. [PMID: 30504220 DOI: 10.1074/jbc.ra118.006111] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/27/2018] [Indexed: 01/05/2023] Open
Abstract
Imidazole-containing dipeptides (IDPs), such as carnosine and anserine, are found exclusively in various animal tissues, especially in the skeletal muscles and nerves. IDPs have antioxidant activity because of their metal-chelating and free radical-scavenging properties. However, the underlying mechanisms that would fully explain IDP antioxidant effects remain obscure. Here, using HPLC-electrospray ionization-tandem MS analyses, we comprehensively investigated carnosine and its related small peptides in the soluble fractions of mouse tissue homogenates and ubiquitously detected 2-oxo-histidine-containing dipeptides (2-oxo-IDPs) in all examined tissues. We noted enhanced production of the 2-oxo-IDPs in the brain of a mouse model of sepsis-associated encephalopathy. Moreover, in SH-SY5Y human neuroblastoma cells stably expressing carnosine synthase, H2O2 exposure resulted in the intracellular production of 2-oxo-carnosine, which was associated with significant inhibition of the H2O2 cytotoxicity. Notably, 2-oxo-carnosine showed a better antioxidant activity than endogenous antioxidants such as GSH and ascorbate. Mechanistic studies indicated that carnosine monooxygenation is mediated through the formation of a histidyl-imidazole radical, followed by the addition of molecular oxygen. Our findings reveal that 2-oxo-IDPs are metal-catalyzed oxidation products present in vivo and provide a revised paradigm for understanding the antioxidant effects of the IDPs.
Collapse
Affiliation(s)
- Hideshi Ihara
- Department of Biological Science, Graduate School of Science, Sakai, Osaka 599-8531, Japan.
| | - Yuki Kakihana
- Department of Biological Science, Graduate School of Science, Sakai, Osaka 599-8531, Japan
| | - Akane Yamakage
- Department of Biological Science, Graduate School of Science, Sakai, Osaka 599-8531, Japan
| | - Kenji Kai
- Department of Biological Chemistry, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka 599-8531, Japan
| | - Takahiro Shibata
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-0054, Japan; Japan Agency for Medical Research and Development, CREST, Tokyo 102-0076, Japan
| | - Koji Uchida
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; Japan Agency for Medical Research and Development, CREST, Tokyo 102-0076, Japan; Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 113-8657, Japan.
| |
Collapse
|
4
|
Zhong KL, Lu MY, Liu F, Mei Y, Zhang XJ, Zhang H, Zan J, Sun XO, Tan W. Isosteviol Sodium Protects Neural Cells Against Hypoxia-Induced Apoptosis Through Inhibiting MAPK and NF-κB Pathways. J Stroke Cerebrovasc Dis 2018; 28:175-184. [PMID: 30539754 DOI: 10.1016/j.jstrokecerebrovasdis.2018.09.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/11/2018] [Accepted: 09/15/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Stevioside, isolated from the herb Stevia rebaudiana, has been widely used as a food sweetener all over the world. Isosteviol Sodium (STV-Na), an injectable formulation of isosteviol sodium salt, has been proved to possess much greater solubility and bioavailability and exhibit protective effects against cerebral ischemia injury in vivo by inhibiting neuron apoptosis. However, the underlying mechanisms of the neuroprotective effects STV-Na are still not completely known. In the present study, we investigated the effects of STV-Na on neuronal cell death caused by hypoxia in vitro and its underlying mechanisms. METHODS We used cobalt chloride (CoCl2) to expose mouse neuroblastoma N2a cells to hypoxic conditions in vitro. RESULTS Our results showed that pretreatment with STV-Na (20 μM) significantly attenuated the decrease of cell viability, lactate dehydrogenase release and cell apoptosis under conditions of CoCl2-induced hypoxia. Meanwhile, STV-Na pretreatment significantly attenuated the upregulation of intracellular Ca2+ concentration and reactive oxygen species production, and inhibited mitochondrial depolarization in N2a cells under conditions of CoCl2-induced hypoxia. Furthermore, STV-Na pretreatment significantly downregulated expressions of nitric oxide synthase, interleukin-1β, tumor necrosis factor-α, interleukin-6, nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) signalings in N2a cells under conditions of CoCl2-induced hypoxia. CONCLUSIONS Taken together, STV-Na protects neural cells against hypoxia-induced apoptosis through inhibiting MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Kai-Lun Zhong
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Min-Yi Lu
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Fei Liu
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Ying Mei
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xue-Ju Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Hao Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Jie Zan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xiao-Ou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Wen Tan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
5
|
Huang J, Wang T, Yu D, Fang X, Fan H, Liu Q, Yi G, Yi X, Liu Q. l-Homocarnosine attenuates inflammation in cerebral ischemia-reperfusion injury through inhibition of nod-like receptor protein 3 inflammasome. Int J Biol Macromol 2018; 118:357-364. [PMID: 29890246 DOI: 10.1016/j.ijbiomac.2018.06.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 10/14/2022]
Abstract
We investigated the therapeutic effects of l-homocarnosine against inflammation in a rat model of cerebral ischemia-reperfusion injury. Rats were grouped into control, middle cerebral artery occlusion (MCAO), 0.5 mM l-homocarnosine + MCAO, and 1 mM l-homocarnosine + MCAO treatment groups. Superoxide dismutase (SOD), glutathione peroxidase (Gpx), catalase, lipid peroxidation, and reduced glutathione (GSH) levels were measured. Neurological scores were assessed, and histopathology, scanning electron microscopy (SEM), and fluorescence microscopy analyses were conducted. The mRNA expression levels of nod-like receptor protein 3 (NLRP3), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6) and protein expression levels of NLRP3 were assessed. l-Homocarnosine supplementation substantially increased SOD, catalase, Gpx, and GSH levels, whereas it reduced the levels of lipid peroxidation relative to MCAO rats. l-Homocarnosine significantly reduced the infarct area and neurological deficit score, as well as histopathological alteration, apoptosis, and necrosis in brain tissue. The mRNA expression levels of NLRP3, TNF-α, and IL-6 were increased in MCAO rats, whereas l-homocarnosine supplementation reduced mRNA expression by >40%, and NLRP3 protein expression was reduced by >30% in 1 mM l-homocarnosine-treated MCAO rats. We propose that l-homocarnosine exerts a protective effect in cerebral ischemia-reperfusion injury-induced rats by downregulating NLRP3 expression.
Collapse
Affiliation(s)
- Jing Huang
- Department of Pharmacology, Hainan Medical University, Haikou, Hainan 571199, China
| | - Tao Wang
- International Nursing School, Hainan Medical University, Haikou, Hainan 571199, China
| | - Daorui Yu
- Department of Pharmacology, Hainan Medical University, Haikou, Hainan 571199, China
| | - Xingyue Fang
- Department of Pharmacology, Hainan Medical University, Haikou, Hainan 571199, China
| | - Haofei Fan
- Department of Pharmacology, Hainan Medical University, Haikou, Hainan 571199, China
| | - Qiang Liu
- Department of Pharmacology, Hainan Medical University, Haikou, Hainan 571199, China
| | - Guohui Yi
- Department of Pharmacology, Hainan Medical University, Haikou, Hainan 571199, China
| | - Xinan Yi
- Department of Anatomy, School of Basic Medicine and Life Science, Hainan Medical University, Haikou, Hainan 571199, China
| | - Qibing Liu
- Department of Pharmacology, Hainan Medical University, Haikou, Hainan 571199, China.
| |
Collapse
|
6
|
Neuroprotective Effects of Bioactive Compounds and MAPK Pathway Modulation in "Ischemia"-Stressed PC12 Pheochromocytoma Cells. Brain Sci 2018; 8:brainsci8020032. [PMID: 29419806 PMCID: PMC5836051 DOI: 10.3390/brainsci8020032] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/24/2018] [Accepted: 02/02/2018] [Indexed: 02/08/2023] Open
Abstract
This review surveys the efforts taken to investigate in vitro neuroprotective features of synthetic compounds and cell-released growth factors on PC12 clonal cell line temporarily deprived of oxygen and glucose followed by reoxygenation (OGD/R). These cells have been used previously to mimic some of the properties of in vivo brain ischemia-reperfusion-injury (IRI) and have been instrumental in identifying common mechanisms such as calcium overload, redox potential, lipid peroxidation and MAPKs modulation. In addition, they were useful for establishing the role of certain membrane penetrable cocktails of antioxidants as well as potential growth factors which may act in neuroprotection. Pharmacological mechanisms of neuroprotection addressing modulation of the MAPK cascade and increased redox potential by natural products, drugs and growth factors secreted by stem cells, in either undifferentiated or nerve growth factor-differentiated PC12 cells exposed to ischemic conditions are discussed for future prospects in neuroprotection studies.
Collapse
|
7
|
Ji H, Xu L, Wang Z, Fan X, Wu L. Effects of thymosin β4 on oxygen‑glucose deprivation and reoxygenation‑induced injury. Int J Mol Med 2018; 41:1749-1755. [PMID: 29328391 DOI: 10.3892/ijmm.2018.3369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 01/04/2018] [Indexed: 11/06/2022] Open
Abstract
Cerebral ischemia causes severe brain injury and results in selective neuronal death through programmed cell death mechanisms, including apoptosis and autophagy. Minimizing neuronal injury has been considered a hot topic among clinicians. The present study elucidated the effect of thymosin β4 (Tβ4) on neuronal death induced by cerebral ischemia/reperfusion in PC12 cells that were subjected to oxygen‑glucose deprivation and reoxygenation (OGD/R). The survival, apoptotic and autophagy rates of PC12 cells were investigated. Tβ4 pre‑conditioning prior to OGD/R was performed to evaluate PC12‑cell viability and the protective mechanisms of Tβ4. Tβ4 significantly increased cell survival after OGD/R. Tβ4 inhibited the release of lactate dehydrogenase, downregulated malondialdehyde and upregulated the activities of glutathione peroxidase and superoxide dismutase. In addition, Tβ4 attenuated OGD/R‑associated decreases in the expression of P62 and the anti‑apoptotic protein B‑cell lymphoma‑2, as well as the upregulation of autophagy mediators, including autophagy‑related protein‑5 and the ratio of microtubule‑associated protein 1 light chain 3 (LC3) II vs. LC3 I. These results suggested that Tβ4 effectively inhibits cell apoptosis and autophagy induced by OGD/R. To the best of our knowledge, the present study was the first to report on the antioxidant, anti‑apoptotic and anti‑autophagic effects of Tβ4 in neuronal‑like PC12 cells. These results suggested that Tβ4 may be explored as a potential treatment for cerebral ischemia.
Collapse
Affiliation(s)
- Hua Ji
- Department of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Linhao Xu
- Department of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Zheng Wang
- Department of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Xinli Fan
- Department of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Lihui Wu
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
8
|
Hung CT, Chen LD, Hou CW. Neuroprotection of a sesamin derivative, 1, 2-bis [(3-methoxyphenyl) methyl] ethane-1, 2-dicaroxylic acid (MMEDA) against ischemic and hypoxic neuronal injury. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:1324-1330. [PMID: 29238467 PMCID: PMC5722992 DOI: 10.22038/ijbms.2017.9543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Objective(s): Stroke may cause severe neuronal damage. The sesamin have been demonstrated to possess neuroprotection by its antioxidant and anti-inflammatory properties. One sesamin derivative was artificially composited, 1, 2-bis [(3-methoxyphenyl) methyl] ethane-1, 2-dicaroxylic acid (MMEDA) had been developed to study its antioxidative activity and neuroprotection. Materials and Methods: The infaction of Sprague Dawley (SD) rats and hypoxia models of BV-2 microglia or PC12 cells were investigated for in vivo and in vitro test respectively. Lipid peroxidation and reactive oxygen species (ROS), prostaglandin E2 (PGE2) and related signaling pathways from hypoxic cells were analyzed by ELISA or Western blot assay, respectively. Results: MMEDA showed a protective effect when given 90 min after the focal cerebral ischemia. The neuroprotection of MMEDA was further confirmed by attenuating ROS and PGE2 release from hypoxic BV-2 or PC12 cells. MMEDA significantly reduced hypoxia-induced JNK and caspase-3 (survival and apoptotic pathways) in PC12 cells. Conclusion: The neuroprotective effect of MMEDA on ischemia/hypoxia models was involved with its antioxidative activity and anti-inflammatory effects. These results suggest that MMEDA exert effective neuroprotection against ischemia/hypoxia injury.
Collapse
Affiliation(s)
- Chang-Tsen Hung
- Department of Health and Leisure Management Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Li-Dian Chen
- Department of Convalescence Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou China
| | - Chien-Wei Hou
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| |
Collapse
|
9
|
Screening and identification of neuroprotective compounds produced by Lactobacillus paracasei subsp. paracasei NTU 101. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.07.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
10
|
Pavlin M, Repič M, Vianello R, Mavri J. The Chemistry of Neurodegeneration: Kinetic Data and Their Implications. Mol Neurobiol 2015; 53:3400-3415. [PMID: 26081152 DOI: 10.1007/s12035-015-9284-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/03/2015] [Indexed: 01/01/2023]
Abstract
We collected experimental kinetic rate constants for chemical processes responsible for the development and progress of neurodegeneration, focused on the enzymatic and non-enzymatic degradation of amine neurotransmitters and their reactive and neurotoxic metabolites. A gross scheme of neurodegeneration on the molecular level is based on two pathways. Firstly, reactive species oxidise heavy atom ions, which enhances the interaction with alpha-synuclein, thus promoting its folding to the beta form and giving rise to insoluble amyloid plaques. The latter prevents the function of vesicular transport leading to gradual neuronal death. In the second pathway, radical species, OH(·) in particular, react with the methylene groups of the apolar part of the lipid bilayer of either the cell or mitochondrial wall, resulting in membrane leakage followed by dyshomeostasis, loss of resting potential and neuron death. Unlike all other central neural system (CNS)-relevant biogenic amines, dopamine and noradrenaline are capable of a non-enzymatic auto-oxidative reaction, which produces hydrogen peroxide. This reaction is not limited to the mitochondrial membrane where scavenging enzymes, such as catalase, are located. On the other hand, dopamine and its metabolites, such as dopamine-o-quinone, dopaminechrome, 5,6-dihydroxyindole and indo-5,6-quinone, also interact directly with alpha-synuclein and reversibly inhibit plaque formation. We consider the role of the heavy metal ions, selected scavengers and scavenging enzymes, and discuss the relevance of certain foods and food supplements, including curcumin, garlic, N-acetyl cysteine, caffeine and red wine, as well as the long-term administration of non-steroid anti-inflammatory drugs and occasional tobacco smoking, that could all act toward preventing neurodegeneration. The current analysis can be employed in developing strategies for the prevention and treatment of neurodegeneration, and, hopefully, aid in the building of an overall kinetic molecular model of neurodegeneration itself.
Collapse
Affiliation(s)
- Matic Pavlin
- Computational Biophysics, German Research School for Simulation Sciences, Joint Venture of RWTH Aachen University and Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Computational Biomedicine, Institute for Advanced Simulations (IAS-5/INM-9), 52425, Jülich, Germany
| | - Matej Repič
- Laboratory of Computational Chemistry and Biochemistry, Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Robert Vianello
- Quantum Organic Chemistry Group, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000, Zagreb, Croatia.
| | - Janez Mavri
- National Institute of Chemistry, Hajdrihova 19, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
11
|
Vogel KR, Ainslie GR, Jansen EEW, Salomons GS, Gibson KM. Torin 1 partially corrects vigabatrin-induced mitochondrial increase in mouse. Ann Clin Transl Neurol 2015; 2:699-706. [PMID: 26125044 PMCID: PMC4479529 DOI: 10.1002/acn3.200] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 12/30/2022] Open
Abstract
Recent findings in mice with targeted deletion of the GABA-metabolic enzyme succinic semialdehyde dehydrogenase revealed a new role for supraphysiological GABA (4-aminobutyric acid) in the activation of the mechanistic target of rapamycin (mTOR) that results in disruption of endogenous mitophagy. Employing biochemical and electron microscopic methodology, we examined the hypothesis that similar outcomes would be observed during intervention with vigabatrin, whose antiepileptic capacity hinges on central nervous system GABA elevation. Vigabatrin intervention was associated with significantly enhanced mitochondrial numbers and areas in normal mice that could be selectively normalized with the rapalog and mechanistic target of rapamycin inhibitor, Torin 1. Moreover, short-term administration of vigabatrin induced apoptosis and enhanced phosphorylation of mechanistic target of rapamycin Ser 2448 in liver. Our results provide new insight into adverse outcomes associated with vigabatrin intervention, and the first evidence that its administration is associated with increased mitochondrial number in central and peripheral tissues that may associate with mechanistic target of rapamycin function and enhanced cell death.
Collapse
Affiliation(s)
- Kara R Vogel
- Section of Experimental and Systems Pharmacology, College of Pharmacy, Washington State University Spokane, Washington
| | - Garrett R Ainslie
- Section of Experimental and Systems Pharmacology, College of Pharmacy, Washington State University Spokane, Washington
| | - Erwin E W Jansen
- Metabolic Unit, Department of Clinical Chemistry, VU University Medical Center Neuroscience Campus, Amsterdam, The Netherlands
| | - Gajja S Salomons
- Metabolic Unit, Department of Clinical Chemistry, VU University Medical Center Neuroscience Campus, Amsterdam, The Netherlands
| | - K Michael Gibson
- Section of Experimental and Systems Pharmacology, College of Pharmacy, Washington State University Spokane, Washington
| |
Collapse
|
12
|
Lahiani A, Zahavi E, Netzer N, Ofir R, Pinzur L, Raveh S, Arien-Zakay H, Yavin E, Lazarovici P. Human PLacental eXpanded (PLX) mesenchymal-like adherent stromal cells confer neuroprotection to nerve growth factor (NGF)-differentiated PC12 cells exposed to ischemia by secretion of IL-6 and VEGF. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:422-30. [DOI: 10.1016/j.bbamcr.2014.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 11/03/2014] [Accepted: 11/10/2014] [Indexed: 12/21/2022]
|
13
|
Guo H, Kong S, Chen W, Dai Z, Lin T, Su J, Li S, Xie Q, Su Z, Xu Y, Lai X. Apigenin Mediated Protection of OGD-Evoked Neuron-Like Injury in Differentiated PC12 Cells. Neurochem Res 2014; 39:2197-210. [DOI: 10.1007/s11064-014-1421-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 08/05/2014] [Accepted: 08/19/2014] [Indexed: 12/17/2022]
|
14
|
Protective effect of vitexin compound B-1 against hypoxia/reoxygenation-induced injury in differentiated PC12 cells via NADPH oxidase inhibition. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2014; 387:861-71. [DOI: 10.1007/s00210-014-1006-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 06/06/2014] [Indexed: 10/25/2022]
|
15
|
Protective effect of a sesamin derivative, 3-bis (3-methoxybenzyl) butane-1, 4-diol on ischemic and hypoxic neuronal injury. J Biomed Sci 2014; 21:15. [PMID: 24548760 PMCID: PMC3975964 DOI: 10.1186/1423-0127-21-15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 02/17/2014] [Indexed: 01/24/2023] Open
Abstract
Background Stroke is one of the leading causes of neuronal death. Sesamin is known for neuroprotection by its antioxidant and anti-inflammatory properties but it lacks blood–brain barrier (BBB) activity. A panel of sesamin derivatives was screened and 3-bis (3-methoxybenzyl) butane-1,4-diol (BBD) was selected for high BBB activity and tested for its neuroprotective effect. Methods The focal cerebral ischemia of Sprague–Dawley rats and hypoxia models of murine BV-2 microglia or PC12 cells under oxygen/glucose deprivation were used for in vivo and in vitro test, respectively. Lipid peroxidation and superoxide dismutase (SOD) activity from the ischemic brain were tested and reactive oxygen species (ROS), cytokine production, prostaglandin (PGE2) and related signaling pathways from hypoxic cells were examined by ELISA or Western blot assay, respectively. Results BBD showed a protective effect when given 90 min after the focal cerebral ischemia. It also reduced lipid peroxidation and preserved SOD activity from the ischemic brain. The mechanism of BBD was further confirmed by attenuating ROS, cytokine production, and PGE2 release from hypoxic BV-2 or PC12 cells. BBD significantly reduced hypoxia-induced c-Jun N-terminal kinases (JNK) and modulated AKT-1 and caspase-3 (survival and apoptotic pathways) in BV-2 cells, and inhibited hypoxia-induced JNK and cyclooxygenase-2 activation in PC12 cells. Conclusions The neuroprotective effect of BBD on ischemia/hypoxia models was involved with antioxidant and anti-inflammatory effects. The result would help the development of new CNS drug for protection of ischemia/hypoxia injury.
Collapse
|
16
|
Attanasio F, Convertino M, Magno A, Caflisch A, Corazza A, Haridas H, Esposito G, Cataldo S, Pignataro B, Milardi D, Rizzarelli E. Carnosine inhibits Aβ(42) aggregation by perturbing the H-bond network in and around the central hydrophobic cluster. Chembiochem 2013; 14:583-92. [PMID: 23440928 DOI: 10.1002/cbic.201200704] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Indexed: 02/06/2023]
Abstract
Aggregation of the amyloid-β peptide (Aβ) into fibrillar structures is a hallmark of Alzheimer's disease. Thus, preventing self-assembly of the Aβ peptide is an attractive therapeutic strategy. Here, we used experimental techniques and atomistic simulations to investigate the influence of carnosine, a dipeptide naturally occurring in the brain, on Aβ aggregation. Scanning force microscopy, circular dichroism and thioflavin T fluorescence experiments showed that carnosine does not modify the conformational features of Aβ42 but nonetheless inhibits amyloid growth. Molecular dynamics (MD) simulations indicated that carnosine interacts transiently with monomeric Aβ42 by salt bridges with charged side chains, and van der Waals contacts with residues in and around the central hydrophobic cluster ((17)LVFFA(21)). NMR experiments on the nonaggregative fragment Aβ12-28 did not evidence specific intermolecular interactions between the peptide and carnosine, in agreement with MD simulations. However, a close inspection of the spectra revealed that carnosine interferes with the local propensity of the peptide to form backbone hydrogen bonds close to the central hydrophobic cluster (residues E22, S26 and N27). Finally, MD simulations of aggregation-prone Aβ heptapeptide segments show that carnosine reduces the propensity to form intermolecular backbone hydrogen bonds in the region 18-24. Taken together, the experimental and simulation results (cumulative MD sampling of 0.2 ms) suggest that, despite the inability of carnosine to form stable contacts with Aβ, it might block the pathway toward toxic aggregates by perturbing the hydrogen bond network near residues with key roles in fibrillogenesis.
Collapse
Affiliation(s)
- Francesco Attanasio
- Istituto di Biostrutture e Bioimmagini-UOS CT, Consiglio Nazionale delle Ricerche, V.le A. Doria 6, 95125 Catania, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Tang Y, Cui YC, Wang XJ, Wu AL, Hu GF, Luo FL, Sun JK, Sun J, Wu LK. Neural progenitor cells derived from adult bone marrow mesenchymal stem cells promote neuronal regeneration. Life Sci 2012; 91:951-8. [PMID: 23000028 DOI: 10.1016/j.lfs.2012.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 08/18/2012] [Accepted: 09/11/2012] [Indexed: 01/17/2023]
Abstract
AIM It is well known that neural stem/progenitor cells (NS/PC) are an ideal cell type for the treatment of central nervous system (CNS) disease. However, ethical problems have severely hampered fetal NS/PC from being widely used as a source for stem cell therapy. Recently, it has been demonstrated that autologous bone marrow mesenchymal stem cells (BMSC) can transdifferentiate into neural progenitor cells (NPC). The biological function of BMSC derived NPC (MDNPC) in neuronal systems remains unknown. In the present study, we aimed to investigate whether MDNPC can promote in vitro neural regeneration, a process comprising mainly the generation of neurons and neurotransmitters. MAIN METHODS We co-cultured BMSC, MDNPC or fetal NS/PC with PC12 cells and studied their roles on proliferation, neurite formation and dopamine release from PC12 cells. Furthermore, we also explored the mechanisms by which MDNPC regulate dopamine secretion from PC12 derived neural cells using Western blot. KEY FINDINGS We found that both MDNPC and NS/PC had similar morphologies and there were no significant differences between MDNPC and NS/PC in promoting PC12 cell proliferation, neurite outgrowth, and dopamine release. We also demonstrated that NS/PC induced dopamine secretion was associated with an upregulation of dopamine transporter (DAT) levels. SIGNIFICANCE In summary, MDNPC were comparable to NS/PC in promoting neural regeneration, indicating that MDNPC are a promising candidate source of neural stem cells for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yue Tang
- State Key Laboratory of Translational Cardiovascular Medicine, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gundimeda U, McNeill TH, Elhiani AA, Schiffman JE, Hinton DR, Gopalakrishna R. Green tea polyphenols precondition against cell death induced by oxygen-glucose deprivation via stimulation of laminin receptor, generation of reactive oxygen species, and activation of protein kinase Cε. J Biol Chem 2012; 287:34694-708. [PMID: 22879598 DOI: 10.1074/jbc.m112.356899] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
As the development of synthetic drugs for the prevention of stroke has proven challenging, utilization of natural products capable of preconditioning neuronal cells against ischemia-induced cell death would be a highly useful complementary approach. In this study using an oxygen-glucose deprivation and reoxygenation (OGD/R) model in PC12 cells, we show that 2-day pretreatment with green tea polyphenols (GTPP) and their active ingredient, epigallocatechin-3-gallate (EGCG), protects cells from subsequent OGD/R-induced cell death. A synergistic interaction was observed between GTPP constituents, with unfractionated GTPP more potently preconditioning cells than EGCG. GTPP-induced preconditioning required the 67-kDa laminin receptor (67LR), to which EGCG binds with high affinity. 67LR also mediated the generation of reactive oxygen species (ROS) via activation of NADPH oxidase. An exogenous ROS-generating system bypassed 67LR to induce preconditioning, suggesting that sublethal levels of ROS are indeed an important mediator in GTPP-induced preconditioning. This role for ROS was further supported by the fact that antioxidants blocked GTPP-induced preconditioning. Additionally, ROS induced an activation and translocation of protein kinase C (PKC), particularly PKCε from the cytosol to the membrane/mitochondria, which was also blocked by antioxidants. The crucial role of PKC in GTPP-induced preconditioning was supported by use of its specific inhibitors. Preconditioning was increased by conditional overexpression of PKCε and decreased by its knock-out with siRNA. Collectively, these results suggest that GTPP stimulates 67LR and thereby induces NADPH oxidase-dependent generation of ROS, which in turn induces activation of PKC, particularly prosurvival isoenzyme PKCε, resulting in preconditioning against cell death induced by OGD/R.
Collapse
Affiliation(s)
- Usha Gundimeda
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, USA
| | | | | | | | | | | |
Collapse
|
19
|
Lecht S, Rotfeld E, Arien‐Zakay H, Tabakman R, Matzner H, Yaka R, Lelkes PI, Lazarovici P. Neuroprotective effects of nimodipine and nifedipine in the NGF‐differentiated PC12 cells exposed to oxygen‐glucose deprivation or trophic withdrawal. Int J Dev Neurosci 2012; 30:465-9. [DOI: 10.1016/j.ijdevneu.2012.05.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 05/26/2012] [Accepted: 05/26/2012] [Indexed: 02/08/2023] Open
Affiliation(s)
- Shimon Lecht
- Integrated Cellular Tissue Engineering & Regenerative Medicine LaboratorySchool of Biomedical Engineering, Science and Health SystemsDrexel UniversityPhiladelphiaPA19102USA
| | - Elena Rotfeld
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Hadar Arien‐Zakay
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Rinat Tabakman
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Henry Matzner
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Rami Yaka
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| | - Peter I. Lelkes
- Integrated Cellular Tissue Engineering & Regenerative Medicine LaboratorySchool of Biomedical Engineering, Science and Health SystemsDrexel UniversityPhiladelphiaPA19102USA
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug ResearchFaculty of MedicineThe Hebrew University of JerusalemJerusalem91120Israel
| |
Collapse
|
20
|
Kritis A, Pourzitaki C, Klagas I, Chourdakis M, Albani M. Proteases inhibition assessment on PC12 and NGF treated cells after oxygen and glucose deprivation reveals a distinct role for aspartyl proteases. PLoS One 2011; 6:e25950. [PMID: 22028798 PMCID: PMC3196512 DOI: 10.1371/journal.pone.0025950] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 09/14/2011] [Indexed: 11/24/2022] Open
Abstract
Hypoxia is a severe stressful condition and induces cell death leading to neuronal loss both to the developing and adult nervous system. Central theme to cellular death is the activation of different classes of proteases such as caspases calpains and cathepsins. In the present study we investigated the involvement of these proteases, in the hypoxia-induced PC12 cell death. Rat PC12 is a model cell line for experimentation relevant to the nervous system and several protocols have been developed for either lethal hypoxia (oxygen and glucose deprivation OGD) or ischemic preconditioning (IPS). Nerve Growth Factor (NGF) treated PC12 differentiate to a sympathetic phenotype, expressing neurites and excitability. Lethal hypoxia was established by exposing undifferentiated and NGF-treated PC12 cells to a mixture of N2/CO2 (93:5%) in DMEM depleted of glucose and sodium pyruvate for 16 h. The involvement of caspases, calpains and lysosomal cathepsins D and E to the cell death induced by lethal OGD was investigated employing protease specific inhibitors such as z-VAD-fmk for the caspases, MDL28170 for the calpains and pepstatin A for the cathepsins D and E. Our findings show that pepstatin A provides statistically significant protection from cell death of both naive and NGF treated PC12 cells exposed to lethal OGD. We propose that apart from the established processes of apoptosis and necrosis that are integral components of lethal OGD, the activation of cathepsins D and E launches additional cell death pathways in which these proteases are key partners.
Collapse
Affiliation(s)
- Aristidis Kritis
- Laboratory of Physiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | | | | | | | | |
Collapse
|
21
|
|
22
|
Ban K, Kozar RA. Glutamine protects against apoptosis via downregulation of Sp3 in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2010; 299:G1344-53. [PMID: 20884886 PMCID: PMC3006244 DOI: 10.1152/ajpgi.00334.2010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glutamine plays a key role in intestinal growth and maintenance of gut function, and as we have shown protects the postischemic gut (Kozar RA, Scultz SG, Bick RJ, Poindexter BJ, Desoigne R, Weisbrodt NW, Haber MM, Moore FA. Shock 21: 433-437, 2004). However, the precise mechanisms of the gut protective effects of glutamine have not been well elucidated. In the present study, RNA microarray was performed to obtain differentially expressed genes in intestinal epithelial IEC-6 cells following either 2 mM or 10 mM glutamine. The result demonstrated that specificity protein 3 (Sp3) mRNA expression was downregulated 3.1-fold. PCR and Western blot confirmed that Sp3 expression was decreased by glutamine in a time- and dose-dependent fashion. To investigate the role of Sp3, Sp3 gene siRNA silencing was performed and apoptosis was assessed. Silencing of Sp3 demonstrated a significant increase in Bcl-2 and decrease in Bax protein expression, as well as a decrease in caspase-3, -8, and -9 protein expression and activity. The protein expression of apoptosis-related proteins after hypoxia/reoxygenation was similar to that of normoxia and correlated with a decrease in DNA fragmentation. Importantly, the addition of glutamine to Sp3-silenced cells did not further lessen apoptosis, suggesting that Sp3 plays a major role in the inhibitory effect of glutamine on apoptosis. This novel finding may explain in part the gut-protective effects of glutamine.
Collapse
Affiliation(s)
- Kechen Ban
- Department of Surgery, University of Texas Medical School, Houston, 77030, USA.
| | | |
Collapse
|
23
|
Nakajima Y, Shimazawa M, Mishima S, Hara H. Neuroprotective effects of Brazilian green propolis and its main constituents against oxygen-glucose deprivation stress, with a gene-expression analysis. Phytother Res 2010; 23:1431-8. [PMID: 19277960 DOI: 10.1002/ptr.2797] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Our purpose was to investigate the neuroprotective effects (and the underlying mechanism) exerted by water extract of Brazilian green propolis (WEP) and its main constituents against the neuronal damage induced by oxygen-glucose deprivation (OGD)/reoxygenation in retinal ganglion cells (RGC-5, a rat ganglion cell-line transformed using E1A virus). Cell damage was induced by OGD 4 h plus reoxygenation 18 h exposure. In RGC-5, and also in PC12 (rat pheochromocytoma, neuronal cells), WEP and some of its main constituents attenuated the cell damage. At the end of the period of OGD/reoxygenation, RNA was extracted and DNA microarray analysis was performed to examine the gene-expression profile in RGC-5. Expression of casein kinase 2 (CK2) was down-regulated and that of Bcl-2-related ovarian killer protein (Bok) was up-regulated following OGD stress, results that were confirmed by quantitative reverse transcriptase-PCR (qRT-PCR). These effects were normalized by WEP. Our findings indicate that WEP has neuroprotective effects against OGD/reoxygenation-induced cell damage and that certain constituents of WEP (caffeoylquinic acid derivatives, artepillin C, and p-coumaric acid) may be partly responsible for its neuroprotective effects. Furthermore, the protective mechanism may involve normalization of the expressions of antioxidant- and apoptosis-related genes (such as CK2 and Bok, respectively).
Collapse
Affiliation(s)
- Yoshimi Nakajima
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | | | | | | |
Collapse
|
24
|
Minocycline attenuates both OGD-induced HMGB1 release and HMGB1-induced cell death in ischemic neuronal injury in PC12 cells. Biochem Biophys Res Commun 2009; 385:132-6. [DOI: 10.1016/j.bbrc.2009.04.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2009] [Accepted: 04/14/2009] [Indexed: 12/21/2022]
|
25
|
Pekcetin C, Kiray M, Ergur BU, Tugyan K, Bagriyanik HA, Erbil G, Baykara B, Camsari UM. Carnosine attenuates oxidative stress and apoptosis in transient cerebral ischemia in rats. ACTA BIOLOGICA HUNGARICA 2009; 60:137-48. [PMID: 19584023 DOI: 10.1556/abiol.60.2009.2.1] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cerebral ischemia leads to cognitive decline and neuronal damage in the hippocampus. Reactive oxygen species (ROS) play an important role in the neuronal loss after cerebral ischemia and reperfusion injury. Carnosine has both antioxidant and neuroprotective effects against ROS. In the present study, the effects of carnosine on oxidative stress, apoptotic neuronal cell death and spatial memory following transient cerebral ischemia in rats were investigated. Transient ischemia was induced by occlusion of right common carotid artery of rats for 30 min and reperfusion for 24 h or 1 week. Rats received intraperitoneal injection of 250 mg/kg carnosine or saline 30 min prior to experiment. Determination of antioxidant enzyme activities was performed spectrophotometrically. To detect apoptotic cells, TUNEL staining was performed using an In Situ Cell Death Detection Kit. Carnosine treatment elicited a significant decrease in lipid peroxidation and increase in antioxidant enzyme activities in ischemic rat brains. The number of TUNEL-positive cells was decreased significantly in carnosine-treated group when compared with the ischemia-induction group. Carnosine treatment did not provide significant protection from ischemia induced deficits in spatial learning. The results show that carnosine is effective as a prophylactic treatment for brain tissue when it is administered before ischemia without affecting spatial memory.
Collapse
Affiliation(s)
- C Pekcetin
- Department of Histology & Embryology, Dokuz Eylul University Medical School, Balcova, Izmir 35340, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Hipkiss AR. Carnosine and its possible roles in nutrition and health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2009; 57:87-154. [PMID: 19595386 DOI: 10.1016/s1043-4526(09)57003-9] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The dipeptide carnosine has been observed to exert antiaging activity at cellular and whole animal levels. This review discusses the possible mechanisms by which carnosine may exert antiaging action and considers whether the dipeptide could be beneficial to humans. Carnosine's possible biological activities include scavenger of reactive oxygen species (ROS) and reactive nitrogen species (RNS), chelator of zinc and copper ions, and antiglycating and anticross-linking activities. Carnosine's ability to react with deleterious aldehydes such as malondialdehyde, methylglyoxal, hydroxynonenal, and acetaldehyde may also contribute to its protective functions. Physiologically carnosine may help to suppress some secondary complications of diabetes, and the deleterious consequences of ischemic-reperfusion injury, most likely due to antioxidation and carbonyl-scavenging functions. Other, and much more speculative, possible functions of carnosine considered include transglutaminase inhibition, stimulation of proteolysis mediated via effects on proteasome activity or induction of protease and stress-protein gene expression, upregulation of corticosteroid synthesis, stimulation of protein repair, and effects on ADP-ribose metabolism associated with sirtuin and poly-ADP-ribose polymerase (PARP) activities. Evidence for carnosine's possible protective action against secondary diabetic complications, neurodegeneration, cancer, and other age-related pathologies is briefly discussed.
Collapse
Affiliation(s)
- Alan R Hipkiss
- School of Clinicial and Experimental Medicine, College of Medical and Dental Sciences, The Univeristy of Birmingham, Edgbaston, Birmingham, UK
| |
Collapse
|
27
|
Kim KA, Choi HJ, Kim BG, Park YR, Kim JS, Ryu JH, Soh YJ. Hexane Soluble Fraction of Chungpesagan-tang Exhibits Protective Effect against Hypoxia/Reoxygenation-Induced N2a Cell Damage. Biomol Ther (Seoul) 2008. [DOI: 10.4062/biomolther.2008.16.4.377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
28
|
Neurotropic effect of exogenous L-carnosine in cultured slices of the olfactory cortex from rat brain. Bull Exp Biol Med 2008; 146:1-3. [DOI: 10.1007/s10517-008-0227-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
29
|
Arien-Zakay H, Lecht S, Bercu MM, Tabakman R, Kohen R, Galski H, Nagler A, Lazarovici P. Neuroprotection by cord blood neural progenitors involves antioxidants, neurotrophic and angiogenic factors. Exp Neurol 2008; 216:83-94. [PMID: 19070617 DOI: 10.1016/j.expneurol.2008.11.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2008] [Revised: 11/06/2008] [Accepted: 11/14/2008] [Indexed: 12/31/2022]
Abstract
Human umbilical cord blood (HUCB) is a valuable source for cell therapy since it confers neuroprotection in stroke animal models. However, the responsible sub-populations remain to be established and the mechanisms involved are unknown. To explore HUCB neuroprotective properties in a PC12 cell-based ischemic neuronal model, we used an HUCB mononuclear-enriched population of collagen-adherent cells, which can be differentiated in vitro into a neuronal phenotype (HUCBNP). Upon co-culture with insulted-PC12 cells, HUCBNP conferred approximately 30% neuroprotection, as evaluated by decreased lactate dehydrogenase and caspase-3 activities. HUCBNP decreased by 95% the level of free radicals in the insulted-PC12 cells, in correlation with the appearance of antioxidants, as measured by changes in the oxidation-reduction potential of the medium using cyclic-voltammetry. An increased level of nerve growth factor (NGF), vascular endothelial growth factor and basic fibroblast growth factor in the co-culture medium was temporally correlated with a -medium neuroprotection effect, which was partially abolished by heat denaturation. HUCBNP-induced neuroprotection was correlated with changes in gene expression of these neurotrophic factors, while blocked by K252a, an antagonist of the TrkA/NGF receptor. These findings indicate that HUCBNP-induced neuroprotection involves antioxidant(s) and neurotrophic factors, which, by paracrine and/or autocrine interactions between the insulted-PC12 and the HUCBNP cells, conferred neuroprotection.
Collapse
Affiliation(s)
- Hadar Arien-Zakay
- Department of Pharmacology and Experimental Therapeutics, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
30
|
TABAKMAN RINAT, JIANG HAO, SHAHAR IRIS, ARIEN-ZAKAY HADAR, LEVINE ROBERTA, LAZAROVICI PHILIP. Neuroprotection by NGF in the PC12 In Vitro OGD Model. Ann N Y Acad Sci 2008. [DOI: 10.1111/j.1749-6632.2005.tb00013.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
31
|
Focus on cyclo(His-Pro): history and perspectives as antioxidant peptide. Amino Acids 2007; 35:283-9. [DOI: 10.1007/s00726-007-0629-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Accepted: 10/30/2007] [Indexed: 10/22/2022]
|
32
|
Chung H, Kim E, Lee DH, Seo S, Ju S, Lee D, Kim H, Park S. Ghrelin inhibits apoptosis in hypothalamic neuronal cells during oxygen-glucose deprivation. Endocrinology 2007; 148:148-59. [PMID: 17053024 DOI: 10.1210/en.2006-0991] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Ghrelin is an endogenous ligand for the GH secretagogue receptor, produced and secreted mainly from the stomach. Ghrelin stimulates GH release and induces positive energy balances. Previous studies have reported that ghrelin inhibits apoptosis in several cell types, but its antiapoptotic effect in neuronal cells is unknown. Therefore, we investigated the role of ghrelin in ischemic neuronal injury using primary hypothalamic neurons exposed to oxygen-glucose deprivation (OGD). Here we report that treatment of hypothalamic neurons with ghrelin inhibited OGD-induced cell death and apoptosis. Exposure of neurons to ghrelin caused rapid activation of ERK1/2. Ghrelin-induced activation of ERK1/2 and the antiapoptotic effect of ghrelin were blocked by chemical inhibition of MAPK, phosphatidylinositol 3 kinase, protein kinase C, and protein kinase A. Ghrelin attenuated OGD-induced activation of c-Jun NH2-terminal kinase and p-38 but not ERK1/2. We also investigated ghrelin regulation of apoptosis at the mitochondrial level. Ghrelin protected cells from OGD insult by inhibiting reactive oxygen species generation and stabilizing mitochondrial transmembrane potential. In addition, ghrelin-treated cells showed an increased Bcl-2/Bax ratio, prevention of cytochrome c release, and inhibition of caspase-3 activation. Finally, in vivo administration of ghrelin significantly reduced infarct volume in an animal model of ischemia. Our data indicate that ghrelin may act as a survival factor that preserves mitochondrial integrity and inhibits apoptotic pathways.
Collapse
Affiliation(s)
- Hyunju Chung
- Department of Pharmacology, Kyunghee University School of Medicine, Dongdaemun-ku, Seoul 130-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Sheng WW, Li CT, Zhang WP, Yuan YM, Hu H, Fang SH, Zhang L, Wei EQ. Distinct roles of CysLT1 and CysLT2 receptors in oxygen glucose deprivation-induced PC12 cell death. Biochem Biophys Res Commun 2006; 346:19-25. [PMID: 16756959 DOI: 10.1016/j.bbrc.2006.05.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2006] [Accepted: 05/04/2006] [Indexed: 12/29/2022]
Abstract
Cysteinyl leukotrienes are involved in ischemic brain injury, and their receptors (CysLT(1) and CysLT(2)) have been cloned. To clarify which subtype mediates the ischemic neuronal injury, we performed permanent transfection to increase CysLT(1) and CysLT(2) receptor expressions in PC12 cells. Oxygen glucose deprivation (OGD)-induced cell death was detected by Hoechst 33258 and propidium iodide fluorescent staining as well as by flow cytometry. OGD induced late phase apoptosis mainly and necrosis minimally. Over-expression of CysLT(1) receptor decreased and over-expression of CysLT(2) receptor increased OGD-induced cell death. An agonist LTD(4) (10(-7)M) also induced apoptosis, especially in CysLT(2) receptor over-expressing cells. A selective CysLT(1) receptor antagonist montelukast did not affect OGD-induced apoptosis; while non-selective CysLT receptor antagonist Bay u9773 inhibited OGD-induced apoptosis, especially in CysLT(2) receptor over-expressing cells. Thus, CysLT(1) and CysLT(2) receptors play distinct roles in OGD-induced PC12 cell death; CysLT(1) attenuates while CysLT(2) facilitates the cell death.
Collapse
Affiliation(s)
- Wen-Wen Sheng
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310031, China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Petroff OAC, Hyder F, Rothman DL, Mattson RH. Brain Homocarnosine and Seizure Control of Patients Taking Gabapentin or Topiramate. Epilepsia 2006; 47:495-8. [PMID: 16529611 DOI: 10.1111/j.1528-1167.2006.00457.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE To assess the relation between seizure control and brain homocarnosine and gamma-aminobutyric acid (GABA) levels of patients with complex partial seizures taking gabapentin (GBP) or topiramate (TPM) as adjunctive therapy. METHODS In vivo measurements of GABA and homocarnosine were made of a 14-cc volume in the occipital cortex by using (1)H spectroscopy with a 2.1-Tesla magnetic resonance spectrometer and an 8-cm surface coil. Poor seizure control was defined as more recent seizures than the median for the two groups of patients studied. RESULTS Homocarnosine levels were higher in patients with better seizure control than in those with poor control. No differences were found in the intracellular GABA levels between the patients who responded to GBP or TPM compared with those who did not. CONCLUSIONS In the visual neocortex, which is remote from the presumed seizure-onset zone, higher homocarnosine levels were associated with better seizure control in the patients taking GBP or TPM as adjunctive therapy; elevated intracellular GABA levels appeared to offer no additional protection.
Collapse
Affiliation(s)
- Ognen A C Petroff
- Department of Neurology, Yale University, New Haven, Connecticut 06520-8018, USA.
| | | | | | | |
Collapse
|
35
|
Nakajima T, Wakasa T, Okuma Y, Inanami O, Nomura Y, Kuwabara M, Kawahara K. Dual inhibition of protein phosphatase-1/2A and calpain rescues nerve growth factor-differentiated PC12 cells from oxygen-glucose deprivation-induced cell death. J Neurosci Res 2006; 83:459-68. [PMID: 16385561 DOI: 10.1002/jnr.20740] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the present study, we examined how the cell survival signaling via cyclic AMP-responsive element binding protein (CREB) and Akt, and the cell death signaling via cystein proteases, calpain and caspase-3, are involved in oxygen-glucose deprivation (OGD) followed by reoxygenation (OGD/reoxygenation)-induced cell death in nerve growth factor (NGF)-differentiated PC12 cells. OGD/reoxygenation-induced cell death was evaluated by LDH release into the culture medium. The level of LDH release was low (9.0% +/- 4.1%) immediately after 4 hr of OGD (0 hr of reoxygenation), was significantly increased to 28.6% +/- 6.6% at 3 hr of reoxygenation, and remained at similar levels at 6 and 20 hr of reoxygenation, suggesting that reoxygenation at least for 3 hr resulted in the loss of cell membrane integrity. After 4 hr of OGD followed by 3 hr of reoxygenation, dephosphorylation of phosphorylated CREB (pCREB), but not phosphorylated Akt (pAkt), was induced. Under these conditions, calpain- but not caspase-3-mediated alpha-spectrin breakdown product was increased, indicating that OGD/reoxygenation also induced an increase in calpain activity. The restoration of pCREB by protein phosphatase (PP)-1/2A inhibitors or the inhibition of excessive activation of calpain by calpain inhibitor did not reduce OGD/reoxygenation-induced LDH release. Cotreatment with PP-1/2A and calpain inhibitors reduced OGD/reoxygenation-induced LDH release. The present study suggests that a balance in the phosphorylation and proteolytic signaling is involved in the survival of NGF-differentiated PC12 cells.
Collapse
Affiliation(s)
- Takayuki Nakajima
- Laboratory of Cellular Cybernetics, Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Minelli A, Bellezza I, Grottelli S, Pinnen F, Brunetti L, Vacca M. Phosphoproteomic analysis of the effect of cyclo-[His-Pro] dipeptide on PC12 cells. Peptides 2006; 27:105-13. [PMID: 16137790 DOI: 10.1016/j.peptides.2005.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2005] [Revised: 07/12/2005] [Accepted: 07/12/2005] [Indexed: 12/23/2022]
Abstract
The effects of dipeptide cyclo-[His-Pro] (CHP), known to participate in the appetite behavior and food intake control, have been investigated using PC12 cells in culture as model system. We found that only in the presence of experimental conditions that cause cellular stress the cyclic dipeptide affect cellular proliferation and protects from apoptosis. It greatly enhances the phosphorylation of hsp27, alpha-B-crystallin, Cdc2, and p-38 MAPK, whereas it decreases the phosphorylation of MEK1, Cav 2, GSK3a, PKB/Akt, PKCdelta, PKCgamma, and Erk2. PKA and PKG are involved in ERK1/2 deactivation via a receptor that appears to be dually coupled to Gs and Gq protein subfamilies.
Collapse
Affiliation(s)
- Alba Minelli
- Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Università di Perugia, via del Giochetto, 06123 Perugia, Italy.
| | | | | | | | | | | |
Collapse
|
37
|
Lortie K, Huang D, Chakravarthy B, Comas T, Hou ST, Lin-Chao S, Morley P. The gas7 protein potentiates NGF-mediated differentiation of PC12 cells. Brain Res 2005; 1036:27-34. [PMID: 15725398 DOI: 10.1016/j.brainres.2004.12.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Revised: 11/27/2004] [Accepted: 12/04/2004] [Indexed: 12/26/2022]
Abstract
The growth-arrest-specific protein gas7 is required for morphological differentiation of cultured mouse cerebellar neurons and PC12 cells. Moreover, its overexpression in various cell types induces neurite-like outgrowth. The role of gas7 in neuronal differentiation was further characterized by adenovirus-mediated overexpression in PC12 cells and quantification of the expression of various neuronal markers, in the absence and presence of different concentrations of nerve growth factor (NGF). The potential neuroprotective activity of gas7 against various neurotoxic insults was also assessed. In addition to promoting the formation of neurite-like extensions, overexpression of gas7 potentiated NGF-mediated neuronal differentiation of PC12 cells, as shown by the enhanced expression of the neuronal proteins betaIII-tubulin, synaptotagmin, alpha7 subunit of the acetylcholine receptor, and dihydropyrimidinase related protein-3. This effect was exerted independently of cell cycle progression, as gas7 did not affect proliferation of PC12 cells. While some differentiation enhancers protect PC12 cells against lethal insults, gas7 overexpression in PC12 cells did not protect against oxygen-glucose deprivation, the calcium ionophore A23187, or the nitric oxide donor sodium nitroprusside, suggesting that gas7 is not neuroprotective. The ability of gas7 to potentiate neuronal differentiation makes it a potential therapeutic target to promote re-establishment of neuronal connections in the injured or diseased brain, such as following stroke.
Collapse
Affiliation(s)
- Karine Lortie
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|