1
|
Haridevamuthu B, Raj D, Kesavan D, Muthuraman S, Kumar RS, Mahboob S, Al-Ghanim KA, Almutairi BO, Arokiyaraj S, Gopinath P, Arockiaraj J. Trihydroxy piperlongumine protects aluminium induced neurotoxicity in zebrafish: Behavioral and biochemical approach. Comp Biochem Physiol C Toxicol Pharmacol 2023; 268:109600. [PMID: 36889534 DOI: 10.1016/j.cbpc.2023.109600] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
Aluminium (Al) is proven to be a potent environmental neurotoxin involved in progressive neurodegeneration. Al primarily induces oxidative stress by free radical generation in the brain, followed by neuronal apoptosis. Antioxidants are promising therapeutic options for Al toxicity. Piperlongumine is traditionally long known for its medicinal properties. Therefore, the present study has been designed to explore the antioxidant role of trihydroxy piperlongumine (THPL) against Al-induced neurotoxicity in the zebrafish model. Zebrafish exposed to AlCl3 exhibited higher oxidative stress and altered locomotion. Adult fish displayed anxiety comorbid with depression phenotype. THPL increases antioxidant enzyme activity by quenching Al-induced free radicals and lipid peroxidation, thus minimizing oxidative damage in the brain. THPL rescues behavior deficits and improves anxiety-like phenotype in adult fish. Histological alterations caused by Al were also attenuated on administration with THPL. Results of the study demonstrate the neuroprotective role of THPL against Al-induced oxidative damage and anxiety, which could be exploited as a psychopharmacological drug.
Collapse
Affiliation(s)
- B Haridevamuthu
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chennai, Tamil Nadu, India. https://twitter.com/haridevamuthub
| | - David Raj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chennai, Tamil Nadu, India
| | - D Kesavan
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chennai, Tamil Nadu, India
| | - Subramani Muthuraman
- Chemistry Division, School of Advanced Sciences, VIT University Chennai Campus, Chennai 600 127, Tamil Nadu, India
| | - Rajendran Saravana Kumar
- Chemistry Division, School of Advanced Sciences, VIT University Chennai Campus, Chennai 600 127, Tamil Nadu, India
| | - Shahid Mahboob
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Khalid Abdullah Al-Ghanim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Bader O Almutairi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Selvaraj Arokiyaraj
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Pushparathinam Gopinath
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Chennai, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603 203, Chennai, Tamil Nadu, India.
| |
Collapse
|
2
|
Laminarin Alleviates the Ischemia/Reperfusion Injury in PC12 Cells via Regulation of PTEN/PI3K/AKT Pathway. ADVANCES IN POLYMER TECHNOLOGY 2022. [DOI: 10.1155/2022/9999339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objective. To investigate the protective effect of laminarin on PC12 cells damaged by oxygen glucose deprivation/reoxygenation (OGD/R) and its molecular mechanism. Methods. PC12 cells in the logarithmic phase were randomly divided into the control group, OGD/R group, and OGD/R+laminarin (0.5, 2.5, and 5 μg/ml) group. CCK-8 activity assay kit was used to detect cell viability. ELISA kit was performed to examine the levels of proinflammatory factors (TNF-α, IL-1β, and IL-6) and oxidative stress markers (ROS, LDH, and MPO). In addition, flow cytometry was employed to determine cell cycle and apoptosis. The expression of cell proliferation-related proteins (PCNA and Ki67), apoptosis-related proteins (Bcl-2, Bax, and Caspase-3), and PTEN/PI3K/AKT pathway-related proteins was evaluated by Western blot. Results. Compared with the control group, the cell viability was decreased significantly in the OGD/R group. CCK-8 results showed that laminarin could attenuate the damage of PC12 cell viability induced by OGD/R in a concentration-dependent manner. Meanwhile, the highest concentration of 5 μg/ml laminarin could significantly promote the viability of PC12 cells and the expression of PCNA and Ki67 than the OGD/R group. Additionally, ELISA assays showed that laminarin significantly inhibited the expression of proinflammatory factors (TNF-α, IL-1β, and IL-6) and the levels of oxidative stress markers (ROS, LDH, and MPO). Flow cytometry results demonstrated that laminarin promoted the cell cycle. And laminin upregulated the expression of apoptotic protein Bcl-2, while downregulated the expression of apoptotic proteins Bax and Caspase-3. Finally, laminarin significantly suppressed the expression of PTEN and facilitated the expression of PI3K and p-AKT compared to the OGD/R group. Conclusion. Laminarin could alleviate the OGD/R-induced PC12 cell neuronal injury via promoting cell activity and cycle and inhibiting inflammation, oxidative stress, and apoptosis. The mechanism may be related to the downregulation of PTEN protein and the activation of the PI3K/AKT pathway.
Collapse
|
3
|
Effect of Cinepazide Maleate on Serum Inflammatory Factors of ICU Patients with Severe Cerebral Hemorrhage after Surgery. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6562140. [PMID: 34745295 PMCID: PMC8568541 DOI: 10.1155/2021/6562140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/15/2021] [Indexed: 12/01/2022]
Abstract
Objective To explore the effect of cinepazide maleate on serum inflammatory factors of intensive care unit (ICU) patients with severe cerebral hemorrhage after surgery. Methods 116 ICU patients with severe cerebral hemorrhage treated in Taian Maternal and Child Health Hospital from June 2018 to June 2020 were selected as the research objects and randomly divided into the control group and experimental group, with 58 patients in each group. The control group was given routine treatment, while the experimental group was additionally given an intravenous drip of cinepazide maleate to compare the clinical efficacy and serum inflammatory factors between the two groups. Results The total effective rate in the experimental group was higher than that in the control group (P < 0.05). After treatment, the Glasgow Coma Scale (GCS), National Institutes of Health Stroke Scale (NIHSS), and Fugl-Meyer scores in both groups were better than those before treatment, and the scores in the experimental group were better than those in the control group (P < 0.05). The oxidative stress indexes such as total antioxidant capacity (T-Aoc), superoxide dismutase (SOD), and glutathione peroxidase (GSH-PX) in the experimental group were higher than those in the control group, while malondialdehyde (MDA) in the experimental group was lower than that in the control group (P < 0.05). The high-sensitivity C-reactive protein (hsCRP), interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor-α (TNF-α) levels in the experimental group were lower than those in the control group (P < 0.05). Compared with the control group, the cerebrovascular function in the experimental group was significantly improved (P < 0.05), with statistically significant differences. Conclusion Cinepazide maleate can effectively reduce the serum inflammatory factor levels of ICU patients with severe cerebral hemorrhage after surgery, alleviate the oxidative stress response in the body, and improve the cerebrovascular function and cerebral nerve function, which is worthy of clinical promotion.
Collapse
|
4
|
Zhu P, Qian J, Xu Z, Meng C, Zhu W, Ran F, Zhang W, Zhang Y, Ling Y. Overview of piperlongumine analogues and their therapeutic potential. Eur J Med Chem 2021; 220:113471. [PMID: 33930801 DOI: 10.1016/j.ejmech.2021.113471] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/29/2021] [Accepted: 04/10/2021] [Indexed: 01/18/2023]
Abstract
Natural products have long been an important source for discovery of new drugs to treat human diseases. Piperlongumine (PL) is an amide alkaloid isolated from Piper longum L. (long piper) and other piper plants and has received widespread attention because of its diverse biological activities. A large number of PL derivatives have been designed, synthesized and assessed in many pharmacological functions, including antiplatelet aggregation, neuroprotective activities, anti-diabetic activities, anti-inflammatory activities, anti-senolytic activities, immune activities, and antitumor activities. Among them, the anti-tumor effects and application of PL and its derivatives are most extensively studied. We herein summarize the development of PL derivatives, the structure and activity relationships (SARs), and their therapeutic potential on the treatments of various diseases, especially against cancer. We also discussed the challenges and future directions associated with PL and its derivatives in these indications.
Collapse
Affiliation(s)
- Peng Zhu
- Medical School, Nantong University, Nantong, 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau
| | - Jianqiang Qian
- Medical School, Nantong University, Nantong, 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Zhongyuan Xu
- Medical School, Nantong University, Nantong, 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Chi Meng
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Weizhong Zhu
- Medical School, Nantong University, Nantong, 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Fansheng Ran
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Wei Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau.
| | - Yanan Zhang
- Medical School, Nantong University, Nantong, 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China.
| | - Yong Ling
- Medical School, Nantong University, Nantong, 226001, China; School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China.
| |
Collapse
|
5
|
Meng L, Ma H, Meng J, Li T, Zhu Y, Zhao Q. Costunolide attenuates oxygen‑glucose deprivation/reperfusion‑induced mitochondrial‑mediated apoptosis in PC12 cells. Mol Med Rep 2021; 23:411. [PMID: 33786628 PMCID: PMC8025489 DOI: 10.3892/mmr.2021.12050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
The present study investigated the effect of costunolide (CT), a compound extracted from Aucklandia lappa Decne, to attenuate oxygen-glucose deprivation/reperfusion (OGD/R)-induced mitochondrial-mediated apoptosis in PC12 cells. The present study used molecular docking technology to detect the binding of CT with mitochondrial apoptotic protein targets. A model of oxygen-glucose deprivation for 2 h and reperfusion for 24 h in PC12 cells was used to mimic cerebral ischemic injury. Cell viability and damage were measured using the Cell Counting kit-8 and lactate dehydrogenase (LDH) cytotoxicity assay kits. Cellular apoptosis was analyzed using flow cytometry. A fluorescence microscope determined intracellular [Ca2+] and mitochondrial membrane potential. Furthermore, immunofluorescence and Western blot analyses were used to detect the expression of apoptosis-associated proteins. CT contains binding sites with Caspase-3, Caspase-9 and Caspase-7. CT markedly enhanced cell viability, inhibited LDH leakage, increased intracellular [Ca2+], stabilized the mitochondrial membrane potential, increased the expression of Bcl-2 and inhibited the expression of Apaf-1, Bax, cleaved-caspase-7, cleaved-caspase-9 and cleaved-caspase-3. CT may markedly protect PC12 cells from damage caused by OGD/R, and its mechanism is associated with blocking the calcium channel and inhibiting mitochondrial-mediated apoptosis.
Collapse
Affiliation(s)
- Lanqing Meng
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Huixia Ma
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Jinni Meng
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Tingting Li
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yafei Zhu
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China.,College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Qipeng Zhao
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
6
|
Zhao Z, Song H, Xie J, Liu T, Zhao X, Chen X, He X, Wu S, Zhang Y, Zheng X. Research progress in the biological activities of 3,4,5-trimethoxycinnamic acid (TMCA) derivatives. Eur J Med Chem 2019; 173:213-227. [PMID: 31009908 PMCID: PMC7115657 DOI: 10.1016/j.ejmech.2019.04.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 01/02/2023]
Abstract
TMCA (3,4,5-trimethoxycinnamic acid) ester and amide are privileged structural scaffolds in drug discovery which are widely distributed in natural products and consequently produced diverse therapeutically relevant pharmacological functions. Owing to the potential of TMCA ester and amide analogues as therapeutic agents, researches on chemical syntheses and modifications have been carried out to drug-like candidates with broad range of medicinal properties such as antitumor, antiviral, CNS (central nervous system) agents, antimicrobial, anti-inflammatory and hematologic agents for a long time. At the same time, SAR (structure-activity relationship) studies have draw greater attention among medicinal chemists, and many of the lead compounds were derived for various disease targets. However, there is an urgent need for the medicinal chemists to further exploit the precursor in developing chemical entities with promising bioactivity and druggability. This review concisely summarizes the synthesis and biological activity for TMCA ester and amide analogues. It also comprehensively reveals the relationship of significant biological activities along with SAR studies. 3,4,5-Trimethoxycinnamic acid (TMCA) derivatives show applications in different pathophysiological conditions due to its privileged structural scaffolds. Natural derived TMCA analogues and chemically modified TMCA ester and amide analogues and their bioactivities are focused in this review. Additionally, it also comprehensively summarized the relationship of significant biological activities along with SAR studies of synthetic TMCA derivatives.
Collapse
Affiliation(s)
- Zefeng Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Huanhuan Song
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China; Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Jing Xie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Tian Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Xue Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Xufei Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China
| | - Xirui He
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Shaoping Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China; Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, Shaanxi, 710069, China.
| | - Yongmin Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China; Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, Shaanxi, 710069, China; Sorbonne Université, Institut Parisien de Chimie Moléculaire, CNRS UMR 8232, 4 place Jussieu, 75005, Paris, France
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, 229 Taibai Road, Xi'an, 710069, China.
| |
Collapse
|
7
|
Catalpol Inhibits Ischemia-Induced Premyelinating Oligodendrocyte Damage through Regulation of Intercellular Calcium Homeostasis via Na⁺/Ca 2+ Exchanger 3. Int J Mol Sci 2018; 19:ijms19071925. [PMID: 29966349 PMCID: PMC6073132 DOI: 10.3390/ijms19071925] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 06/15/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
The heightened vulnerability of premyelinating oligodendrocytes (PreOLs) in response to hypoxia⁻ischemia may contribute to perinatal white matter injury and subsequent neurobehavioral dysfunction. Intracellular Ca2+ overload is considered a crucial mechanism predisposing PreOLs to ischemic injury. We previously reported that catalpol, an iridoid glycoside extracted from Rehmannia root, inhibits intracellular Ca2+ overload of PreOLs in an in vitro ischemia model. However, the exact underlying mechanisms remain elusive. In the present study, we aimed to investigate the protective effects of catalpol on PreOLs and to explore the underlying mechanisms involved in the modulation of intracellular Ca2+ homeostasis. Postnatal day 2 (P2) Sprague-Dawley (SD) rats subjected to bilateral common carotid artery ligation followed by exposure to 8% oxygen for 10 min were used as a rat model of neonatal hypoxia⁻ischemia. We found that catalpol significantly improved behavioral functions and prevented PreOL loss and myelination deficit after hypoxia⁻ischemia. Our in vitro studies also confirmed the direct effects of catalpol on oxygen-glucose deprivation (OGD)-induced cell death and arrested maturation of PreOLs. Moreover, we demonstrated that catalpol significantly inhibited intracellular Ca2+ overload and promoted the expression of Na⁺/Ca2+ exchanger 3 (NCX3). Finally, we found that catalpol significantly reduced mitochondrial damage and subsequent extracellular signal-regulated kinase 1/2 (ERK1/2) and poly-ADP-ribose polymerase-1 (PARP-1) activation. Treatment with NCX3-preferring inhibitor 2-[2-[4-(4-nitrobenzyloxy)phenyl]ethyl]isothiourea (KB-R7943) significantly reversed the protective effects of catalpol on PreOLs under OGD. Overall, our data suggest that catalpol protects PreOLs from ischemic injury through regulation of intercellular Ca2+ homeostasis via upregulation of NCX3 activity.
Collapse
|
8
|
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
|
9
|
Hassanzadeh P, Atyabi F, Dinarvand R, Dehpour AR, Azhdarzadeh M, Dinarvand M. Application of nanostructured lipid carriers: the prolonged protective effects for sesamol in in vitro and in vivo models of ischemic stroke via activation of PI3K signalling pathway. Daru 2017; 25:25. [PMID: 29262855 PMCID: PMC5738862 DOI: 10.1186/s40199-017-0191-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/01/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Treatment of the ischemic stroke has remained a major healthcare challenge. The phenolic compound, sesamol, has shown promising antioxidant and neuroprotective effects, however, fast clearance may negatively affect its efficiency. This, prompted us to incorporate sesamol into the nanostructured lipid carriers (S-NLCs) and evaluate its therapeutic potential in in vitro and in vivo models of ischemic stroke. METHODS S-NLCs formulations were prepared by high-pressure homogenization followed by physicochemical characterization, evaluation of the bioactivity of the optimal formulation in oxygen-glucose deprivation (OGD) and global cerebral ischemia/reperfusion (I/R) injury and implication of phosphatidylinositol 3-kinase (PI3K) pathway in this regard. Two- or three-way ANOVA, Mann-Whitney U test, and Student's t-test were used for data analysis. RESULTS Formation of S-NLCs which exhibited a controlled release profile, was confirmed by scanning electron microscope and differential scanning calorimetry. 1- and 8-h OGD followed by 24 h re-oxygenation significantly reduced PC12 cell viability, increased lactate dehydrogenase activity and the number of condensed nuclei, and induced oxidative stress as revealed by increased malondialdehyde level and decreased glutathione content and superoxide dismutase and catalase activities. Sesamol (80 and 100 μM) reduced the cytotoxicity, oxidative stress, and cellular damage only after 1-h OGD, while, S-NLCs (containing 80 and 100 μM of sesamol) were effective at both time points. Intravenous injections of S-NLCs (20 and 25 mg/kg) into rats markedly attenuated I/R-induced neurobehavioural deficits, cellular damage, and oxidative stress, while, free sesamol failed. Pre-treatment with PI3K inhibitor, LY294002, abolished the protective effects against OGD or I/R. CONCLUSIONS S-NLCs improve the pharmacological profile of sesamol and provide longer lasting protective effects for this phenolic phytochemical. This nanoformulation by activating PI3K pathway may serve as a promising candidate for neuroprotection against the cerebral stroke or other neurodegenerative disorders. Sesamol-loaded NLCs, a promising nanoformulation against the ischemic stroke.
Collapse
Affiliation(s)
- Parichehr Hassanzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad-Reza Dehpour
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Azhdarzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Meshkat Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Hassanzadeh P, Arbabi E, Atyabi F, Dinarvand R. Ferulic acid-loaded nanostructured lipid carriers: A promising nanoformulation against the ischemic neural injuries. Life Sci 2017; 193:64-76. [PMID: 29196052 DOI: 10.1016/j.lfs.2017.11.046] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 11/07/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023]
Abstract
AIMS Treatment of the ischemic stroke has remained a major healthcare challenge. The phenolic compound, ferulic acid (FA), has shown promising antioxidant and neuroprotective effects, however, low bioavailability may negatively affect its efficiency. This, prompted us to incorporate FA into the nanostructured lipid carriers (FA-NLCs) and evaluate its therapeutic potential in in vitro and in vivo models of ischemic stroke. MAIN METHODS FA-NLCs were prepared by high-pressure homogenization followed by physicochemical characterization, evaluation of the bioactivity of FA-NLCs in oxygen-glucose deprivation (OGD) and global cerebral ischemia/reperfusion (I/R) injury and implication of phosphatidylinositol 3-kinase (PI3K) pathway in this regard. KEY FINDINGS Formation of FA-NLCs which exhibited a controlled release profile, was confirmed by scanning electron microscope and differential scanning calorimetry. 1- and 8-h OGD followed by 24h re-oxygenation significantly reduced PC12 cell viability, increased lactate dehydrogenase activity and number of condensed nuclei, and induced oxidative stress as revealed by increased malondialdehyde and decreased glutathione content and superoxide dismutase and catalase activities. FA (80 and 100μM) reduced the cytotoxicity, oxidative stress, and cellular damage only after 1-h OGD, while, FA-NLCs (containing 80 and 100μM of FA) were effective at both time points. Intravenous injections of FA-NLCs (20 and 25mg/kg) into rats significantly attenuated I/R-induced neurobehavioural deficits, cellular damage, and oxidative stress, while, FA failed. Pre-treatment with PI3K inhibitor, LY294002, abolished the protective effects against OGD or I/R. SIGNIFICANCE FA-NLCs by improving the pharmacological profile of FA and activating PI3K pathway might be of therapeutic value in cerebral stroke.
Collapse
Affiliation(s)
- Parichehr Hassanzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Elham Arbabi
- Research Center for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Han N, Kim YJ, Park SM, Kim SM, Lee JS, Jung HS, Lee EJ, Kim TK, Kim TN, Kwon MJ, Lee SH, Kim MK, Rhee BD, Park JH. Repeated Glucose Deprivation/Reperfusion Induced PC-12 Cell Death through the Involvement of FOXO Transcription Factor. Diabetes Metab J 2016; 40:396-405. [PMID: 27766247 PMCID: PMC5069396 DOI: 10.4093/dmj.2016.40.5.396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/23/2015] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Cognitive impairment and brain damage in diabetes is suggested to be associated with hypoglycemia. The mechanisms of hypoglycemia-induced neural death and apoptosis are not clear and reperfusion injury may be involved. Recent studies show that glucose deprivation/reperfusion induced more neuronal cell death than glucose deprivation itself. The forkhead box O (FOXO) transcription factors are implicated in the regulation of cell apoptosis and survival, but their role in neuronal cells remains unclear. We examined the role of FOXO transcription factors and the involvement of the phosphatidylinositol 3-kinase (PI3K)/Akt and apoptosis-related signaling pathways in PC-12 cells exposed to repeated glucose deprivation/reperfusion. METHODS PC-12 cells were exposed to control (Dulbecco's Modified Eagle Medium [DMEM] containing 25 mM glucose) or glucose deprivation/reperfusion (DMEM with 0 mM glucose for 6 hours and then DMEM with 25 mM glucose for 18 hours) for 5 days. MTT assay and Western blot analysis were performed for cell viability, apoptosis, and the expression of survival signaling pathways. FOXO3/4',6-diamidino-2-phenylindole staining was done to ascertain the involvement of FOXO transcription factors in glucose deprivation/reperfusion conditions. RESULTS Compared to PC-12 cells not exposed to hypoglycemia, cells exposed to glucose deprivation/reperfusion showed a reduction of cell viability, decreased expression of phosphorylated Akt and Bcl-2, and an increase of cleaved caspase-3 expression. Of note, FOXO3 protein was localized in the nuclei of glucose deprivation/reperfusion cells but not in the control cells. CONCLUSION Repeated glucose deprivation/reperfusion caused the neuronal cell death. Activated FOXO3 via the PI3K/Akt pathway in repeated glucose deprivation/reperfusion was involved in genes related to apoptosis.
Collapse
Affiliation(s)
- Na Han
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Onhospital, Busan, Korea
| | - You Jeong Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Su Min Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Seung Man Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Ji Suk Lee
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Korea
| | - Hye Sook Jung
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Korea
| | - Eun Ju Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Tae Kyoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Tae Nyun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Min Jeong Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Soon Hee Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Mi-kyung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Korea
| | - Byoung Doo Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Jeong Hyun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
- Paik Institute for Clinical Research, Molecular Therapy Lab, Inje University, Busan, Korea
| |
Collapse
|
12
|
Chang R, Zhou R, Qi X, Wang J, Wu F, Yang W, Zhang W, Sun T, Li Y, Yu J. Protective effects of aloin on oxygen and glucose deprivation-induced injury in PC12 cells. Brain Res Bull 2016; 121:75-83. [PMID: 26772628 DOI: 10.1016/j.brainresbull.2016.01.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/28/2015] [Accepted: 01/04/2016] [Indexed: 01/11/2023]
Abstract
The present study aims to determine whether aloin could protect cells from ischemic and reperfusion injury in vitro and to elucidate the related mechanisms. Oxygen and glucose deprivation model in PC12 cells was used in the present study. 2-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), lactate dehydrogenase (LDH) assay and Hoechst 33342 nuclear staining were used to evaluate the protective effects of aloin, at concentrations of 10, 20, or 40 μg/mL in PC12 cells. PCR was applied to detect fluorescence caspase-3, Bax and Bcl-2 mRNA expression in PC12 cells. The contents of malondialdehyde (MDA), superoxide dismutase (SOD) activity were evaluated by biochemical method. The concentration of intracellular-free calcium [Ca(2+)]i, mitochondrial membrane potential (MMP) were determined to estimate the degree of neuronal damage. It was shown that aloin (10, 20, and 40 μg/mL) significantly attenuated PC12 cells damage with characteristics of an increased injured cells absorbance of MTT and releases of LDH, decreasing cell apoptosis, and antagonizing decreases in SOD activity and increase in MDA level induced by OGD-reoxygenation. Meanwhile pretreatment with aloin significantly reduced injury-induced intracellular ROS, increased MMP (P<0.01), but it inhibited [Ca(2+)]i (P<0.01) elevation in a dose-dependent manner. Furthermore, pre-treatment with aloin significantly up-regulated Bcl-2 mRNA expression, down-regulated Bax mRNA expression and consequently activated caspase-3 mRNA expression in a dose-dependent manner. The results indicated that the protection of aloin on OGD-induced apoptosis in PC12 cells is associated with its suppression on OGD-induced oxidative stress and protection on mitochondrial function and inhibition of caspase activity. Alion could be a promising candidate in the development of a novel class of anti-ischemic agent.
Collapse
Affiliation(s)
- Renyuan Chang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Ru Zhou
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Xue Qi
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jing Wang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Fan Wu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Wenli Yang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Wannian Zhang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Ningxia Key Lab of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yuxiang Li
- College of Nursing, Ningxia Medical University, Yinchuan, China.
| | - Jianqiang Yu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China; Ningxia Hui Medicine Modern Engineering Research Center, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
13
|
Liu Y, Zhang RY, Zhao J, Dong Z, Feng DY, Wu R, Shi M, Zhao G. Ginsenoside Rd Protects SH-SY5Y Cells against 1-Methyl-4-phenylpyridinium Induced Injury. Int J Mol Sci 2015; 16:14395-408. [PMID: 26114390 PMCID: PMC4519848 DOI: 10.3390/ijms160714395] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/08/2015] [Accepted: 06/17/2015] [Indexed: 12/24/2022] Open
Abstract
Ginsenoside Rd (GSRd), one of the main active monomer compounds from the medical plant Panaxginseng, has been shown to promote neuronal survival in models of ischemic cerebral damage. As an extending study, here we examined whether GSRd could exert a beneficial effect in an experimental Parkinson disease (PD) model in vitro, in which SH-SY5Y cells were injured by 1-methyl-4-phenylpyridinium (MPP+), an active metabolic product of the classical Parkinsonian toxin1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our results, from the addition of different concentrations of GSRd (1, 10 and 50 μM), showed that GSRd at 1 and 10 μM could significantly attenuate MPP+-induced cell death. This protective effect may be ascribed to its ability to reduce intracellular reactive oxygen species levels, enhance antioxidant enzymatic activities, preserve the activity of respiratory complex I, stabilize the mitochondrial membrane potential and increase intracellular ATP levels. Additionally, the PI3K/Akt survival-signaling pathway was also involved in the protective effect of GSRd. Finally, using a mouse PD model in vivo, we also found that GSRd obviously reversed the loss of tyrosine hydroxylase-positive cells in substanitia nigra induced by MPTP. Thus, our findings demonstrated that GSRd showed a significant neuro-protective effect against experimental PD models, which may involve its antioxidant effects and mitochondrial function preservation.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
- 153rd Hospital of People's Liberation Army, Zhengzhou 450007, China.
| | - Ren-Yu Zhang
- Cadet Brigade of Fourth Military Medical University, Xi'an 710032, China.
| | - Jun Zhao
- 316th Hospital of People's Liberation Army, Beijing 100091, China.
| | - Zheng Dong
- Cadet Brigade of Fourth Military Medical University, Xi'an 710032, China.
| | - Dong-Yun Feng
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Rui Wu
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Ming Shi
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|