101
|
Zhang Y, Liu J, Lv Y, Zhang C, Guo S. LncRNA meg3 suppresses hepatocellular carcinoma in vitro and vivo studies. Am J Transl Res 2019; 11:4089-4099. [PMID: 31396320 PMCID: PMC6684911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/09/2019] [Indexed: 06/10/2023]
Abstract
Although abnormal expression of the long non-coding RNA (lncRNA) MEG3 has been reported in multiple cancer types, the role of MEG3 in the pathobiology of hepatocellular carcinoma (HCC) remains unknown. This study evaluated the expression of lncRNA MEG3 and a microRNA (miRNA-10a-5p) implicated in HCC metastasis in cancer and carcinoma adjacent tissues of HCC samples (n=30 each) via in situ hybridization and quantitative RT-PCR. The effects of overexpressing either MEG3 alone, or MEG3 in combination with miRNA-10a-5p, on the proliferation, apoptosis, cell cycle progression, migration, and invasion of HepG2 cells were evaluated using functional assays. Dual luciferase reporter assays and western blotting were employed to delineate the mechanisms of MEG3 and miRNA-10a-5p regulation of key oncogenes and tumor suppressors in HCC cells. Compared to carcinoma-adjacent regions, MEG3 expression was downregulated in cancer regions of HCC samples; by contrast, miRNA-10a-5p was overexpressed in cancer regions compared to tumor-adjacent areas. Furthermore, overexpression of MEG3 (a) decreased proliferation, migration, and invasion of HepG2 cells; (b) enhanced apoptosis and the proportion of HepG2 cells in G1 of the cell cycle; (c) increased the expression of phosphatase and tensin homolog (PTEN), Bcl2-associated X (Bax), and p53 proteins; and (d) decreased the expression of miRNA-10a-5p, AKT, p-AKT, Bcl-2, and the matrix metalloproteinases (MMPs)-2 and -9. Furthermore, miRNA-10a-5p bound the 3-untranslated region of PTEN mRNA and downregulated PTEN protein expression. Taken together, these data suggest that MEG3 regulates the PTEN/AKT/MMP-2/MMP-9 signaling axis and contributes to HCC development by targeting miRNA-10a-5p.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Hand and Foot Surgery, Linyi Central HospitalLinyi 276400, Shandong, China
| | - Jinfeng Liu
- Department of Oncology, Rizhao Hospital of Traditional Chinese MedicineRizhao 276800, Shandong, China
| | - Yan Lv
- Department of Internal Medicine (4), Shandong Provincial Chest HospitalJinan 250013, Shandong, China
| | - Chao Zhang
- Department of Gastroenterology and Hepatobiliary Discipline, Rizhao Hospital of Traditional Chinese MedicineRizhao 276800, Shandong, China
| | - Shuai Guo
- Department of Oncology, Shandong Provincial Chest HospitalJinan 250013, Shandong, China
| |
Collapse
|
102
|
Belanova A, Beseda D, Chmykhalo V, Stepanova A, Belousova M, Khrenkova V, Gavalas N, Zolotukhin P. Berberine Effects on NFκB, HIF1A and NFE2L2/AP-1 Pathways in HeLa Cells. Anticancer Agents Med Chem 2019; 19:487-501. [DOI: 10.2174/1871520619666181211121405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/19/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022]
Abstract
Background:
Berberine has multitudinous anti-cancer stem cells effects making it a highly promising
candidate substance for the next-generation cancer therapy. However, berberine modes of action predispose it to
significant side-effects that probably limit its clinical testing and application.
Materials and Methods:
HeLa cells were treated with two concentrations of berberine (30 and 100 µM) for 24
hours to assess the functioning of the NFE2L2/AP-1, NFκB and HIF1A pathways using 22 RNAs expression
qPCR-based analysis.
Results:
Berberine effects appeared to be highly dose-dependent, with the lower concentration being capable of
suppressing the NFκB functioning and the higher concentration causing severe signaling side-effects seen in the
HIF1A pathway and the NFE2L2 sub-pathways, and especially and more importantly in the AP-1 sub-pathway.
Conclusion:
The results of the study suggest that berberine has clinically valuable anti-NFκB effects however
jeopardized by its side effects on the HIF1A and especially NFE2L2/AP-1 pathways, its therapeutic window
phenomenon and its cancer type-specificity. These, however, may be ameliorated using the cocktail approach,
provided there is enough data on signaling effects of berberine.
Collapse
Affiliation(s)
- Anna Belanova
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| | - Darya Beseda
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| | - Victor Chmykhalo
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| | - Alisa Stepanova
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| | - Mariya Belousova
- English Language Department for Natural Sciences Faculties, Southern Federal University, 5 Sorge st., 344090, Rostov-on-Don, Russian Federation
| | - Vera Khrenkova
- Rostov State Medical University, 119 Suvorova st., 344022, Rostov-on-Don, Russian Federation
| | - Nikolaos Gavalas
- Division of Clinical Therapeutics, National and Kapodistrian University of Athens, 80 Vas. Sofias Av., 11521, Athens, Greece
| | - Peter Zolotukhin
- Biomedical Innovations LLC, 112 Mechnikova st., 344013, Rostov-on-Don, Russian Federation
| |
Collapse
|
103
|
In Vitro and In Vivo Neuroprotective Effects of Stellettin B Through Anti-Apoptosis and the Nrf2/HO-1 Pathway. Mar Drugs 2019; 17:md17060315. [PMID: 31146323 PMCID: PMC6627894 DOI: 10.3390/md17060315] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/19/2019] [Accepted: 05/24/2019] [Indexed: 12/21/2022] Open
Abstract
Pharmaceutical agents for halting the progression of Parkinson’s disease (PD) are lacking. The current available medications only relieve clinical symptoms and may cause severe side effects. Therefore, there is an urgent need for novel drug candidates for PD. In this study, we demonstrated the neuroprotective activity of stellettin B (SB), a compound isolated from marine sponges. We showed that SB could significantly protect SH-SY5Y cells against 6-OHDA-induced cellular damage by inhibiting cell apoptosis and oxidative stress through PI3K/Akt, MAPK, caspase cascade modulation and Nrf2/HO-1 cascade modulation, respectively. In addition, an in vivo study showed that SB reversed 6-OHDA-induced a locomotor deficit in a zebrafish model of PD. The potential for developing SB as a candidate drug for PD treatment is discussed.
Collapse
|
104
|
Luo R, Liao Z, Song Y, Yin H, Zhan S, Li G, Ma L, Lu S, Wang K, Li S, Zhang Y, Yang C. Berberine ameliorates oxidative stress-induced apoptosis by modulating ER stress and autophagy in human nucleus pulposus cells. Life Sci 2019; 228:85-97. [PMID: 31047897 DOI: 10.1016/j.lfs.2019.04.064] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023]
Abstract
AIM Nucleus pulposus (NP) cell apoptosis induced by oxidative stress is known to be closely involved in the pathogenesis of intervertebral disc (IVD) degeneration. Berberine, a small molecule derived from Rhizoma coptidis, has been found to exert antioxidative activity and preserve cell viability. The present study aims to investigate whether berberine can prevent NP cell apoptosis under oxidative damage and the potential underlying mechanisms. METHODS AND MATERIALS The effects of berberine on IVD degeneration were investigated both in vitro and in vivo. KEY FINDINGS Our results showed that berberine significantly mitigated oxidative stress-decreased cell viability as well as apoptosis in human NP cells. Berberine treatment could attenuate oxidative stress-induced ER stress and autophagy in a concentration-dependent manner. With 4-PBA (ER stress specific inhibitor) and 3-MA (autophagy specific inhibitor) administration, we demonstrated that berberine inhibited oxidative stress-induced apoptosis by modulating the ER stress and autophagy pathway. We also found that the IRE1/JNK pathway was involved in the induction of ER stress-dependent autophagy. With Ca2+ chelator BAPTA-AM utilization, we revealed that oxidative stress-mediated ER stress and autophagy repressed by berberine could be restored by inducing intracellular Ca2+ dysregulation. Furthermore, in vivo study provided evidence that berberine treatment could retard the process of puncture-induced IVD degeneration in a rat model. SIGNIFICANCE Our results indicate that berberine could prevent oxidative stress-induced apoptosis by modulating ER stress and autophagy, thus offering a novel potential pharmacological treatment strategy for IVD degeneration.
Collapse
Affiliation(s)
- Rongjin Luo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhiwei Liao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huipeng Yin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shengfeng Zhan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Saideng Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yukun Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
105
|
Abstract
Mild environmental stress might have beneficial effects in aging by activating maintenance and repair processes in cells and organs. These beneficial stress effects fit to the concept of hormesis. Prominent stressors acting in a hormetic way are physical exercises, fasting, cold and heat. This review will introduce some toxins, which have been found to induce hormetic responses in animal models of aging research. To highlight the molecular signature of these hormetic effects we will depict signaling pathways affected by low doses of toxins on cellular and organismic level. As prominent examples for signaling pathways involved in both aging processes as well as toxin responses, PI3K/Akt/mTOR- and AMPK-signal transduction will be described in more detail. Due to the striking overlap of signaling pathways mediating toxin induced responses and aging processes we propose considering the ability of low doses of toxins to slow down the rate of aging.
Collapse
|
106
|
Zhang C, Li C, Jia X, Wang K, Tu Y, Wang R, Liu K, Lu T, He C. In Vitro and In Vivo Anti-Inflammatory Effects of Polyphyllin VII through Downregulating MAPK and NF-κB Pathways. Molecules 2019; 24:molecules24050875. [PMID: 30832224 PMCID: PMC6429153 DOI: 10.3390/molecules24050875] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Polyphyllin VII (PP7), a steroidal saponin from Paris polyphylla, has been found to exert strong anticancer activity. Little is known about the anti-inflammatory property of PP7. In this study, the anti-inflammatory activity and its underlying mechanisms of PP7 were evaluated in lipopolysaccharide (LPS)-stimulated RAW264.7 cells and in multiple animal models. Methods: The content of nitric oxide (NO) was determined by spectrophotometry. The levels of prostaglandin E2 (PGE2) and cytokines were measured by enzyme-linked immunosorbent assay (ELISA) assay. The mRNA expression of pro-inflammatory genes was determined by qPCR. The total and phosphorylated protein levels were examined by Western blotting. The in vivo anti-inflammatory activities were evaluated by using mouse and zebrafish models. Results: PP7 reduced the production of NO and PGE2 and the protein and mRNA expressions of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) and enzymes (inducible NO synthase [iNOS], cyclooxygenase-2 [COX-2], and Matrix metalloproteinase-9 [MMP-9]) in LPS-induced RAW264.7 cells by suppressing the NF-κB and MAPKs pathways. Notably, PP7 markedly inhibited xylene-induced ear edema and cotton pellet-induced granuloma formation in mice and suppressed LPS and CuSO4-induced inflammation and toxicity in zebrafish embryos. Conclusion: This study demonstrates that PP7 exerts strong anti-inflammatory activities in multiple in vitro and in vivo models and suggests that PP7 is a potential novel therapeutic agent for inflammatory diseases.
Collapse
Affiliation(s)
- Chao Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| | - Chaoying Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Xuejing Jia
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| | - Kai Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| | - Yanbei Tu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| | - Rongchun Wang
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Shandong Provincial Key Laboratory for Biosensor, Biology Institute of Shandong Academy of Sciences, Jinan 250014, China.
| | - Kechun Liu
- Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Shandong Provincial Key Laboratory for Biosensor, Biology Institute of Shandong Academy of Sciences, Jinan 250014, China.
| | - Tao Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
107
|
Shen S, Liao Q, Feng Y, Liu J, Pan R, Lee SMY, Lin L. Myricanol mitigates lipid accumulation in 3T3-L1 adipocytes and high fat diet-fed zebrafish via activating AMP-activated protein kinase. Food Chem 2019; 270:305-314. [DOI: 10.1016/j.foodchem.2018.07.117] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022]
|
108
|
Wang S, Zhang C, Li Y, Li P, Zhang D, Li C. Anti-liver cancer effect and the mechanism of arsenic sulfide in vitro and in vivo. Cancer Chemother Pharmacol 2018; 83:519-530. [PMID: 30542770 DOI: 10.1007/s00280-018-3755-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE This study aimed at investigating the anti-tumor effect of arsenic sulfide (As2S2) against liver cancer both in vivo and in vitro and to elucidate its underlying mechanisms. METHODS Cell viability of the human hepatocellular carcinoma cell lines SMMC-7721, BEL-7402, HepG2 were measured by CCK-8 assay. The effects of As2S2 on cell proliferation and apoptosis of SMMC-7721 cells were investigated using Calcein-AM and PI staining, Hoechst 33258 staining, crystal violet staining, and JC-1 staining. Cell cycle and Annexin V/PI assay were analyzed via flow cytometry. The expression of apoptosis-related proteins, phosphorylation of PI3K and AKT were detected by Western blotting. H22-bearing mice model was established to evaluate the anti-tumor effect of As2S2 in vivo. HE staining, PCNA was observed via immunohistochemistry, and TUNEL assay was used to assess the anti-proliferation and pro-apoptotic effects of As2S2. RESULTS As2S2 significantly inhibited the growth of human hepatoma cells SMMC-7721, BEL-7402 and HepG2. As2S2 inhibited cell proliferation effectively by inducing G0/G1 cell cycle arrest in SMMC-7721 cells. As2S2 could increase Bax/Bcl-2 ratio, decrease mitochondrial membrane potential, promote the release of cytochrome c, increase the levels of cleaved caspase-3 and PARP, indicating that As2S2 induced apoptosis in SMMC-7721 cells via mitochondrial-mediated apoptosis pathway. Further research showed that As2S2 inhibited the PI3K/AKT signaling pathway leading to apoptotic cell death. In addition, As2S2 significantly inhibited tumor growth in H22-bearing mice and induced apoptosis by deactivating PI3K/AKT pathway, which was consistent with the in vitro results. CONCLUSION These findings suggested that As2S2 could induce apoptosis of liver cancer cells in vitro and in vivo, which was related to PI3K/AKT-mediated mitochondrial pathway and may provide a novel promising therapeutic agent for liver cancer treatment.
Collapse
Affiliation(s)
- Shudan Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Chao Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yumei Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Ping Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Dafang Zhang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Chaoying Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
109
|
Liu Y, Wu X, An J, Lv W, Geng Y, Lou T, Zhang Y. Glaucocalyxin B protects against oxygen-glucose-deprivation/reperfusion-induced neuronal injury in PC-12 cells. J Cell Biochem 2018; 120:6137-6144. [PMID: 30304556 DOI: 10.1002/jcb.27901] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/24/2018] [Indexed: 02/01/2023]
Abstract
Oxidative stress has been implicated in the development of cerebral ischemia/reperfusion (I/R) injury. Glaucocalyxin B (GLB), one of five ent-kauranoid diterpenoids, was reported to possess neuroprotective activity. However, the effect of GLB on oxygen-glucose-deprivation/reperfusion (OGD/R)-induced cell injury in PC-12 cells has not been explored. PC-12 cells was treated with various concentrations of GLB (0, 2.5, 5 and 10 μM), and cell viability was detected using the MTT assay. PC-12 cells were pretreated with the indicated concentration of GLB (2.5-10 μM, 2 hours pretreatment), and were maintained under OGD for 3 hours, followed by 24 hours of reoxygenation. Cell viability was assessed using the MTT assay. The levels of superoxide dismutase, malondialdehyde, and glutathione peroxidase were detected using commercially available ELISA Kits. Intracellular reactive oxygen species level was measured using the fluorescent probe 2',7'-dichlorofluorescein diacetate. The levels of Bcl-2, Bax, p-Akt, Akt, p-mTOR, mTOR were detected using Western blot. Our results revealed that GLB significantly protected PC12 cells against OGD/R-induced cell injury. In addition, GLB efficiently inhibited oxidative stress and cell apoptosis in OGD/R-stimulated PC-12 cells. Mechanistic studies revealed that pretreatment with GLB could induce the activation of Akt/mTOR signaling pathway resulting in protection of OGD-treated PC12 cells. In conclusion, our data indicate for the first time that GLB protects against OGD/R-induced neuronal injury in PC-12 cells. The mechanism of the protective effect of GLB is partially associated with activation of the Akt/mTOR signaling pathway. Thus, GLB may be a potential agent for protection against cerebral I/R injury.
Collapse
Affiliation(s)
- Yuxin Liu
- Department of Pharmacy, Huaihe Hospital of Henan University, Kaifeng, China
| | - Xianchuang Wu
- Department of Pharmacy, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jihong An
- Department of Pharmacy, Huaihe Hospital of Henan University, Kaifeng, China
| | - Weiling Lv
- Department of Pharmacy, Huaihe Hospital of Henan University, Kaifeng, China
| | - Yanna Geng
- Department of Pharmacy, Huaihe Hospital of Henan University, Kaifeng, China
| | - Tingting Lou
- Department of Pharmacy, Huaihe Hospital of Henan University, Kaifeng, China
| | - Yongzhou Zhang
- Department of Pharmacy, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
110
|
Hormesis: Path and Progression to Significance. Int J Mol Sci 2018; 19:ijms19102871. [PMID: 30248927 PMCID: PMC6213774 DOI: 10.3390/ijms19102871] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/06/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022] Open
Abstract
This paper tells the story of how hormesis became recognized as a fundamental concept in biology, affecting toxicology, microbiology, medicine, public health, agriculture, and all areas related to enhancing biological performance. This paper assesses how hormesis enhances resilience to normal aging and protects against a broad spectrum of neurodegenerative, cardiovascular, and other diseases, as well as trauma and other threats to health and well-being. This paper also explains the application of hormesis to several neurodegenerative diseases such as Parkinson’s and Huntington’s disease, macrophage polarization and its systematic adaptive protections, and the role of hormesis in enhancing stem cell functioning and medical applications.
Collapse
|
111
|
Kafi Z, Cheshomi H, Gholami O. 7-Isopenthenyloxycoumarin, Arctigenin, and Hesperidin Modify Myeloid Cell Leukemia Type-1 (Mcl-1) Gene Expression by Hormesis in K562 Cell Line. Dose Response 2018; 16:1559325818796014. [PMID: 30224905 PMCID: PMC6136114 DOI: 10.1177/1559325818796014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/17/2018] [Accepted: 07/24/2018] [Indexed: 12/31/2022] Open
Abstract
Hormesis is a new concept in dose–response relationship. Despite of traditional
dose–response curves, there is a low-dose stimulation and a high-dose inhibition
in this case. Hormesis effect in apoptosis induction/inhibition by natural
compounds is reported previously. Here, we searched this effect for myeloid cell
leukemia type-1 (Mcl-1) gene expression by phytochemicals
7-isopenthenyloxycoumarin (7-IP), arctigenin (Arg), and hesperidin (Hsp). For
this purpose, first we tested the cytotoxicity of various doses of these
compounds against K562 leukemia cell lines for different times by
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method. After that
we explored the effect of various doses of these phytochemicals on Mcl-1 gene
expression for different times by real-time polymerase chain reaction method. We
found that these phytochemicals have cytotoxicity against K562 cell line.
Hesperidin is the most cytotoxic agent. We also found that these natural
compounds have hormetic effect on Mcl-1 gene expression. The hormetic model in
Mcl-1 gene expression is overcompensation stimulation. This phenomenon is
reported for the first time. We conclude that 7-IP, Arg, and Hsp are cytotoxic
against K562 cancerous cells and induce/inhibit Mcl-1 gene expression by
hormesis dose–response relationship.
Collapse
Affiliation(s)
- Zahra Kafi
- Cellular and Molecular Research Center, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Hamid Cheshomi
- Cellular and Molecular Research Center, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran.,Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Omid Gholami
- Cellular and Molecular Research Center, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
112
|
Calabrese V, Santoro A, Trovato Salinaro A, Modafferi S, Scuto M, Albouchi F, Monti D, Giordano J, Zappia M, Franceschi C, Calabrese EJ. Hormetic approaches to the treatment of Parkinson's disease: Perspectives and possibilities. J Neurosci Res 2018; 96:1641-1662. [PMID: 30098077 DOI: 10.1002/jnr.24244] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 01/17/2023]
Abstract
Age-related changes in the brain reflect a dynamic interaction of genetic, epigenetic, phenotypic, and environmental factors that can be temporally restricted or more longitudinally present throughout the lifespan. Fundamental to these mechanisms is the capacity for physiological adaptation through modulation of diverse molecular and biochemical signaling occurring from the intracellular to the network-systemic level throughout the brain. A number of agents that affect the onset and progression of Parkinson's disease (PD)-like effects in experimental models exhibit temporal features, and mechanisms of hormetic dose responses. These findings have particular significance since the hormetic dose response describes the amplitude and range of potential therapeutic effects, thereby affecting the design and conduct of studies of interventions against PD (and other neurodegenerative diseases), and may also be important to a broader consideration of hormetic processes in resilient adaptive responses that might afford protection against the onset and/or progression of PD and related disorders.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania.,IBREGENS, Nutraceuticals and Functional Food Biotechnologies Research Associated, University of Catania, Italy
| | - Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania
| | - Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania
| | - Maria Scuto
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania
| | - Ferdaous Albouchi
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania
| | - Daniela Monti
- Department of Experimental, Clinical and Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - James Giordano
- Departments of Neurology and Biochemistry, and Neuroethics Studies Program, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Mario Zappia
- Department of Medical Sciences, Surgical and Advanced Technologies G.F. Ingrassia, Section of Neurosciences, University of Catania, Italy
| | | | - Edward J Calabrese
- Environmental Health Sciences Division, School of Public Health, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
113
|
Gao J, Liu S, Xu F, Liu Y, Lv C, Deng Y, Shi J, Gong Q. Trilobatin Protects Against Oxidative Injury in Neuronal PC12 Cells Through Regulating Mitochondrial ROS Homeostasis Mediated by AMPK/Nrf2/Sirt3 Signaling Pathway. Front Mol Neurosci 2018; 11:267. [PMID: 30104959 PMCID: PMC6077207 DOI: 10.3389/fnmol.2018.00267] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/13/2018] [Indexed: 12/28/2022] Open
Abstract
Oxidative stress-induced neuronal cell damage is a crucial factor in the pathogenesis of mitochondria-associated neurological diseases. Therefore, elimination of overproduction of mitochondrial reactive oxygen species (mtROS) may be a potential strategy for prevention and treatment of neurological diseases. In the present study, the neuroprotective effects of trilobatin (TLB), a novel small molecule monomer derived from Lithocarpus polystachyus Rehd, and its underlying mechanisms were investigated in vitro using hydrogen peroxide (H2O2)-induced oxidative stress model in a neuron-like PC12 cell. The findings revealed that pre-treatment with TLB dramatically concentration-dependently suppressed H2O2-induced PC12 cells damage by enhancing cell viability, repressed reduction of mitochondrial membrane potential (MMP) and decreased mtROS overgeneration, thereby deferring cell apoptosis. Further study demonstrated that TLB not only increased the enzymatic activities of glutathione peroxidase (GPx), isocitrate dehydrogenase 2 (IDH2),superoxide dismutase 2 (SOD2) and deacetylation of SOD2, but also activated silent mating-type information regulation 2 homolog 3 (Sirt3) within the mitochondria and thereby upregulating forkheadboxO3a (FoxO3a), which regulated mitochondrial DNA genes, then led to improving complex I activity and adenosine triphosphate (ATP) synthesis. What’s more, TLB up-regulated p-adenosine monophosphate-activated protein kinase (AMPK) level, the expression of peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α), and ERRα. Intriguingly, TLB failed to mitigate H2O2-induced PC12 injury in the presence of the AMPK inhibitor (Compound C), indicating that the beneficial effects of TLB on the regulation of mtROS homeostasis were reliance on AMPK -Sirt3 signaling pathway. Moreover, TLB also facilitated nuclear factor erythroid 2-related factor 2 (Nrf2) and promoted antioxidant gene expression in turn, and knockdown of Nrf2 by siRNA dramatically reduced the neuroprotective effects of TLB. Notably, AMPK inhibitor abolished the activation of Nrf2 and Sirt3, whereas, knockdown of Nrf2 blocked the upregulation of Sirt3, but it did not affect p-AMPK level. In conclusion, our findings demonstrate that TLB protects against oxidative injury in neuronal PC12 cells through regulating mtROS homeostasis in the first time, which is, at least partly, mediated through the AMPK/Nrf2/Sirt3 signaling pathway.
Collapse
Affiliation(s)
- Jianmei Gao
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Department of Pharmacology, Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Shuang Liu
- Department of Pharmacology, Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Fan Xu
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yuangui Liu
- Department of Pharmacology, Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Chun Lv
- Department of Pharmacology, Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yan Deng
- Department of Pharmacology, Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jingshan Shi
- Department of Pharmacology, Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Qihai Gong
- Department of Pharmacology, Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
114
|
Li M, Zhou F, Xu T, Song H, Lu B. Acteoside protects against 6-OHDA-induced dopaminergic neuron damage via Nrf2-ARE signaling pathway. Food Chem Toxicol 2018; 119:6-13. [PMID: 29906474 DOI: 10.1016/j.fct.2018.06.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 12/30/2022]
Abstract
Acteoside has been reported to have antioxidant and neuroprotective effect, which is a promising therapeutic way in prevention and treatment of Parkinson's disease. The present study was aimed to understand the neuroprotective effect of acteoside and to elucidate its underlying mechanism. 6-hydroxydopamine (6-OHDA)-induced neural damage in zebrafish model was used to study the protective effect of acteoside on Parkinson's disease (PD). Locomotion behavioral test showed that acteoside could prevent 6-OHDA-stimulated movement disorders. Anti-tyrosine hydroxylase (TH) whole-mount immunostaining analysis showed that acteoside could prevent 6-OHDA-induced dopaminergic neuron death. In addition, pretreatment with acteoside could upregulate antioxidative enzymes by activating the Nrf2/ARE signaling pathway in zebrafish. Meanwhile, acteoside was found to be distributed in the brain after intraperitoneal injection into the adult zebrafish, indicating that this compound could penetrate the blood-brain-barrier (BBB). This study demonstrated that acteoside could penetrate BBB and have potential therapeutic value for PD by activating the Nrf2/ARE signaling pathway and attenuating the oxidative stress.
Collapse
Affiliation(s)
- Maiquan Li
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Fei Zhou
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Tao Xu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Huaxin Song
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Baiyi Lu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
115
|
Berberine Protects Human Retinal Pigment Epithelial Cells from Hydrogen Peroxide-Induced Oxidative Damage through Activation of AMPK. Int J Mol Sci 2018; 19:ijms19061736. [PMID: 29895743 PMCID: PMC6032421 DOI: 10.3390/ijms19061736] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/23/2018] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of central vision loss in the elderly with less effective treatment, especially for dry AMD (90% of AMD). Although the etiology of this disease is not well elucidated, increasing evidences indicate that excessive reactive oxygen species (ROS) impairing the physiological functions of retinal pigment epithelium (RPE) cells may be one of the main causes. Therefore, it could be a great strategy to find some drugs that can effectively protect RPE cells from oxidative damage which is desired to treat and slow the process of AMD. In the present study, a well-known traditional Chinese medicine berberine (BBR) was found to suppress hydrogen peroxide (H2O2)-induced oxidative damage in D407 cells, a human RPE cell line. Pre-treatment of D407 cells with BBR significantly suppressed H2O2-induced cell apoptosis by restoring abnormal changes in nuclear morphology, preventing the decline of mitochondrial membrane potential, reducing lactate dehydrogenase release and inhibiting caspase 3/7 activities induced by H2O2. Western blot analysis showed that BBR was able to stimulate the phosphorylation/activation of AMPK in a time- and dose-dependent manner in D407 cells, while treatment of cells with AMPK pathway inhibitor Compound C, or knockdown of the AMPK by specific siRNA blocked the effect of BBR. Similar results were obtained in primary cultured human RPE cells. Taken together, these results demonstrated that BBR was able to protect RPE cells against oxidative stress via the activation of AMPK pathway. Our findings also indicate the potential application of BBR in AMD treatment.
Collapse
|
116
|
Benvenutti R, Marcon M, Reis CG, Nery LR, Miguel C, Herrmann AP, Vianna MRM, Piato A. N-acetylcysteine protects against motor, optomotor and morphological deficits induced by 6-OHDA in zebrafish larvae. PeerJ 2018; 6:e4957. [PMID: 29868300 PMCID: PMC5985760 DOI: 10.7717/peerj.4957] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022] Open
Abstract
Background Parkinson's disease (PD) is the second most common neurodegenerative disorder. In addition to its highly debilitating motor symptoms, non-motor symptoms may precede their motor counterparts by many years, which may characterize a prodromal phase of PD. A potential pharmacological strategy is to introduce neuroprotective agents at an earlier stage in order to prevent further neuronal death. N-acetylcysteine (NAC) has been used against paracetamol overdose hepatotoxicity by restoring hepatic concentrations of glutathione (GSH), and as a mucolytic in chronic obstructive pulmonary disease by reducing disulfide bonds in mucoproteins. It has been shown to be safe for humans at high doses. More recently, several studies have evidenced that NAC has a multifaceted mechanism of action, presenting indirect antioxidant effect by acting as a GSH precursor, besides its anti-inflammatory and neurotrophic effects. Moreover, NAC modulates glutamate release through activation of the cystine-glutamate antiporter in extra-synaptic astrocytes. Its therapeutic benefits have been demonstrated in clinical trials for several neuropsychiatric conditions but has not been tested in PD models yet. Methods In this study, we evaluated the potential of NAC to prevent the damage induced by 6-hydroxydopamine (6-OHDA) on motor, optomotor and morphological parameters in a PD model in larval zebrafish. Results NAC was able to prevent the motor deficits (total distance, mean speed, maximum acceleration, absolute turn angle and immobility time), optomotor response impairment and morphological alterations (total length and head length) caused by exposure to 6-OHDA, which reinforce and broaden the relevance of its neuroprotective effects. Discussion NAC acts in different targets relevant to PD pathophysiology. Further studies and clinical trials are needed to assess this agent as a candidate for prevention and adjunctive treatment of PD.
Collapse
Affiliation(s)
- Radharani Benvenutti
- Programa de Pós-graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Matheus Marcon
- Programa de Pós-graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos G Reis
- Programa de Pós-graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Laura R Nery
- Programa de Pós-graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Camila Miguel
- Programa de Pós-graduação em Zoologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana P Herrmann
- Programa de Pós-graduação em Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Monica R M Vianna
- Programa de Pós-graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Programa de Pós-graduação em Zoologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Angelo Piato
- Programa de Pós-graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Programa de Pós-graduação em Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
117
|
Zhuang W, Li T, Wang C, Shi X, Li Y, Zhang S, Zhao Z, Dong H, Qiao Y. Berberine exerts antioxidant effects via protection of spiral ganglion cells against cytomegalovirus-induced apoptosis. Free Radic Biol Med 2018; 121:127-135. [PMID: 29715550 DOI: 10.1016/j.freeradbiomed.2018.04.575] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 12/20/2022]
Abstract
Cytomegalovirus (CMV) is the leading cause of sensorineural hearing loss (SNHL) in children because of its damage to the cochlea and spiral ganglion cells. Therefore, it has become a top priority to devise new methods to effectively protect spiral ganglion cells from damage. Berberine (BBR) has gained attention for its vast beneficial biological effects through immunomodulation, and its anti-inflammatory and anti-apoptosis properties. However, the effect of BBR on spiral ganglion cells and molecular mechanisms are still unclear. This study aims to investigate whether BBR has an anti-apoptosis effect in CMV-induced apoptosis in cultured spiral ganglion cells and explore the possible mechanism. In this study, TUNEL and MTT assays significantly demonstrated that low doses of BBR did not promote cell apoptosis and they also inhibited the CMV-induced cultured spiral ganglion cell apoptosis. Immunofluorescence and Western blot assays indicated that the anti-apoptosis effect of BBR was related to Nox3. Mitochondrial calcium and Western blot assays revealed that NMDAR1 mediated this anti-apoptosis effect. Our results demonstrated that BBR exerted an anti-apoptosis effect against CMV in cultured spiral ganglion cells, and the mechanism is related to NMDAR1/Nox3-mediated mitochondrial reactive oxygen species (ROS) generation.
Collapse
Affiliation(s)
- Wei Zhuang
- Clinical Hearing Center, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, Jiangsu, China
| | - Ting Li
- Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou 221004, China
| | - Caiji Wang
- The Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xi Shi
- The Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yalan Li
- The Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Shili Zhang
- The Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Zeqi Zhao
- The Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Hongyan Dong
- Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Yuehua Qiao
- Clinical Hearing Center, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, Jiangsu, China; The Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
118
|
Lee J, Song K, Huh E, Oh MS, Kim YS. Neuroprotection against 6-OHDA toxicity in PC12 cells and mice through the Nrf2 pathway by a sesquiterpenoid from Tussilago farfara. Redox Biol 2018; 18:6-15. [PMID: 29890337 PMCID: PMC6041377 DOI: 10.1016/j.redox.2018.05.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 05/30/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress plays a key role in neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Therefore, the nuclear factor-E2-related factor 2 (Nrf2), a key regulator of the antioxidative response, is considered to be important as a therapeutic target for neurodegenerative diseases. We investigated the underlying mechanism of Nrf2-mediated neuroprotective effects against oxidative stress in the PC12 cell line by 7β-(3-ethyl-cis-crotonoyloxy)-1α-(2-methylbutyryloxy)-3,14-dehydro-Z-notonipetranone (ECN), one of the sesquiterpenoids in Farfarae Flos. Pretreatment of PC12 cells with ECN had a protective effect against hydrogen peroxide (H2O2)- or 6-hydroxydopamine (6-OHDA)-induced cytotoxicity. ECN upregulated the ARE-luciferase activity and induced the mRNA expression of Nrf2 and antioxidant enzyme heme oxygenase-1 (HO-1). Knockdown of Nrf2 by small, interfering RNA (siRNA) abrogated the upregulation of HO-1, indicating that ECN had induced HO-1 via the Nrf2 pathway. Pretreatment with the thiol reducing agents, N-acetylcysteine (NAC) or dithiothreitol (DTT), attenuated Nrf2 activation and HO-1 expression. However, the non-thiol reducing antioxidant, Trolox, failed to inhibit HO-1 induction by ECN. These results suggest that ECN may directly interact with Kelch-like ECH-associated protein 1 (Keap1) and modify critical cysteine thiols present in the proteins responsible for Nrf2-mediated upregulation of HO-1. In a 6-OHDA-induced mouse model of PD, administration of ECN ameliorated motor impairments and dopaminergic neuronal damage. Taken together, ECN exerts neuroprotective effects by activating the Nrf2/HO-1 signaling pathway in both PC12 cells and mice. Thus, ECN, as an Nrf2 activator, could be an attractive therapeutic candidate for the neuroprotection or treatment of neurodegenerative diseases. ECN protects PC12 cells against H2O2- or 6-OHDA-induced cell damage. ECN increases HO-1 expression via the nuclear translocation and ARE binding of Nrf2. Activation of the Nrf2/HO-1 pathway contributes to the protective properties of ECN. ECN ameliorates 6-OHDA-induced motor impairments in mice. ECN prevents 6-OHDA-induced loss of dopaminergic neurons in the mouse brain.
Collapse
Affiliation(s)
- Joohee Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Kwangho Song
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Eugene Huh
- Department of Medical Science of Meridian, Graduate School, Kyung Hee University, Seoul 02447, South Korea; Department of Life and Nanopharmaceutical Sciences, Graduate School and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul 02447, South Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate School and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul 02447, South Korea
| | - Yeong Shik Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
119
|
Li H, Tang Z, Chu P, Song Y, Yang Y, Sun B, Niu M, Qaed E, Shopit A, Han G, Ma X, Peng J, Hu M, Tang Z. Neuroprotective effect of phosphocreatine on oxidative stress and mitochondrial dysfunction induced apoptosis in vitro and in vivo: Involvement of dual PI3K/Akt and Nrf2/HO-1 pathways. Free Radic Biol Med 2018; 120:228-238. [PMID: 29559323 DOI: 10.1016/j.freeradbiomed.2018.03.014] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/03/2018] [Accepted: 03/09/2018] [Indexed: 12/15/2022]
Abstract
Methylglyoxal (MGO), an active metabolite of glucose, is observed in high levels in the tissues and blood of diabetic patients. Phosphocreatine (PCr), a high-energy phosphate compound, exhibits a range of pharmacological actions but little is well known of its neuroprotective action. The aim of the present study was to investigate the neuroprotective effects and the possible mechanisms of PCr. Diabetes is closely associated with neurodegenerative diseases, leading not only to the peripheral nervous system (PNS) and but also to central nervous system (CNS) damage. Therefore, we established two rat models of diabetes in vivo induced by MGO and streptozocin (STZ) respectively, while utilized differentiated PC-12 cells in vitro. Treatment of PC-12 cells with PCr markedly attenuated MGO-induced change of viability, apoptosis, accompanied by decreased levels of caspase-3, casapse-9 and Bcl-2/Bax protein ratio. Determination of cellular respiratory function was performed with intact PC-12 cells and homogenized hippocampal neuron tissue of rat. Reactive oxygen species (ROS) generation was assessed by membrane permeable fluorescent probe DCFH-DA. The expressions of Akt, Nrf2 and HO-1 were examined by Western blot. PCr pretreatment significantly reduced oxidative stress-induced high LDH, MDA level, and ROS production of PC-12 cells. PCr pretreatment also significantly decreased mitochondrial dysfunction in vitro and in vivo. In addition, PCr pretreatment increased the expression of p-Akt, Nrf2 and HO-1, and reduced the apoptosis. Moreover, the expression of Cleaved caspase3 was partially increased and the p-Akt, Nrf2 and HO-1 was partially reduced by a PI3K inhibitor (LY294002). While, compared with LY294002 groups, pre-treatment with PCr at the concentrations of 20 mM significantly reduced the expression of Cleaved caspase3 and increased the expression of p-Akt, Nrf2 and HO-1. Molecular docking assay showed that PCr possessed powerful affinity towards to Akt with lower binding energy. In conclusion, the neuroprotective effects of PCr in vitro and in vivo rely on normalizing mitochondrial function and reducing oxidative stress via Akt mediated Nrf2/HO-1 pathway, suggesting that PCr may be a novel therapeutic candidate for the treatment of diabetes-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Hailong Li
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Zhongyuan Tang
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, PR China
| | - Peng Chu
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Yanlin Song
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Ying Yang
- Dalian Medical University, Affiliated Hosp 2, Neurological Intensive Care Un it, Dalian 116027, PR China
| | - Bin Sun
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Mengyue Niu
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Eskandar Qaed
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Abdullah Shopit
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Guozhu Han
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Xiaodong Ma
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Jinyong Peng
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Min Hu
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, PR China.
| | - Zeyao Tang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, PR China.
| |
Collapse
|
120
|
Lin X, Zhang N. Berberine: Pathways to protect neurons. Phytother Res 2018; 32:1501-1510. [DOI: 10.1002/ptr.6107] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 03/03/2018] [Accepted: 04/05/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Xiaorui Lin
- Second Department of Clinical Medicine; China Medical University; No. 77 Puhe Road Shenyang 110122 PR China
| | - Nan Zhang
- Department of Neuroendocrine Pharmacology, School of Pharmacy; China Medical University; No. 77 Puhe Road Shenyang 110122 PR China
| |
Collapse
|
121
|
Elmazoglu Z, Ergin V, Sahin E, Kayhan H, Karasu C. Oleuropein and rutin protect against 6-OHDA-induced neurotoxicity in PC12 cells through modulation of mitochondrial function and unfolded protein response. Interdiscip Toxicol 2018; 10:129-141. [PMID: 30147420 PMCID: PMC6102676 DOI: 10.1515/intox-2017-0019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 12/14/2017] [Indexed: 12/17/2022] Open
Abstract
Parkinson’s disease (PD) is a highly prevalent neurodegenerative disorder, often associated with oxidative stress-induced transcriptional changes in dopaminergic neurons. Phenolic antioxidants, oleuropein (OLE) and rutin (RUT) have attracted a great interest due to their potential to counteract oxidative protein aggregation and toxicity. This study aimed at examining the effects of OLE and RUT against 6-OHDA-induced stress response in rat pheochromocytoma cells. When differentiated PC12 cells were exposed to oxidative stress composer 6-OHDA (100 μM, 8 h), a decreased mitochondrial membrane potential (ΔΨm) was observed along with a significant loss of cell viability and apoptotic nuclear changes. Exposure to 6-OHDA resulted in unfolded protein response (UPR) in differentiated PC12 cells as evidenced by an increased level of endoplasmic reticulum (ER)-localized transmembrane signal transducer IRE1α, adaptive response proteins ATF-4 and proapoptotic transcription factor CHOP. OLE or RUT pretreatment (24 h) at low doses (1–50 μM) protected the differentiated PC12 cells from 6-OHDA-induced cytotoxicity as assessed by increased viability, improved ΔΨm and inhibited apoptosis, whereas relatively high doses of OLE or RUT (>50 μM) inhibited cell growth and proliferation, indicating a typical hormetic effect. In hormetic doses, OLE and RUT up-regulated 6-OHDA-induced increase in IRE1α, ATF-4 and inhibited CHOP, PERK, BIP and PDI. 6-OHDA-activated XBP1 splicing was also inhibited by OLE or RUT. The presented results suggest that neuroprotection against 6-OHDA-induced oxidative toxicity may be attributable to neurohormetic effects of OLE or RUT at low doses through regulating mitochondrial functions, controlling persistent protein misfolding, activating and/or amplificating the adaptive response-related signaling pathways, leading to UPR prosurvival output.
Collapse
Affiliation(s)
- Zubeyir Elmazoglu
- Department of Medical Pharmacology, Cellular Stress Response and Signal Transduction Research Laboratory, Gazi University, Faculty of Medicine, Ankara, Turkey
| | - Volkan Ergin
- Department of Medical Biology, Erzincan University, Faculty of Medicine, Erzincan, Turkey
| | - Ergin Sahin
- Department of Biology, Ankara University, Faculty of Science, Ankara, Turkey
| | - Handan Kayhan
- Department of Hematology, Gazi University, Faculty of Medicine, Ankara, Turkey
| | - Cimen Karasu
- Department of Medical Pharmacology, Cellular Stress Response and Signal Transduction Research Laboratory, Gazi University, Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
122
|
Zeng XS, Geng WS, Jia JJ. Neurotoxin-Induced Animal Models of Parkinson Disease: Pathogenic Mechanism and Assessment. ASN Neuro 2018; 10:1759091418777438. [PMID: 29809058 DOI: 10.1177/1759091418777438if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Parkinson disease (PD) is the second most common neurodegenerative movement disorder. Pharmacological animal models are invaluable tools to study the pathological mechanisms of PD. Currently, invertebrate and vertebrate animal models have been developed by using several main neurotoxins, such as 6-hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, paraquat, and rotenone. These models achieve to some extent to reproduce the key features of PD, including motor defects, progressive loss of dopaminergic neurons in substantia nigra pars compacta, and the formation of Lewy bodies. In this review, we will highlight the pathogenic mechanisms of those neurotoxins and summarize different neurotoxic animal models with the hope to help researchers choose among them accurately and to promote the development of modeling PD.
Collapse
Affiliation(s)
- Xian-Si Zeng
- 1 College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| | - Wen-Shuo Geng
- 1 College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| | - Jin-Jing Jia
- 1 College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| |
Collapse
|
123
|
Zeng XS, Geng WS, Jia JJ. Neurotoxin-Induced Animal Models of Parkinson Disease: Pathogenic Mechanism and Assessment. ASN Neuro 2018; 10:1759091418777438. [PMID: 29809058 PMCID: PMC5977437 DOI: 10.1177/1759091418777438] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 04/12/2018] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) is the second most common neurodegenerative movement disorder. Pharmacological animal models are invaluable tools to study the pathological mechanisms of PD. Currently, invertebrate and vertebrate animal models have been developed by using several main neurotoxins, such as 6-hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, paraquat, and rotenone. These models achieve to some extent to reproduce the key features of PD, including motor defects, progressive loss of dopaminergic neurons in substantia nigra pars compacta, and the formation of Lewy bodies. In this review, we will highlight the pathogenic mechanisms of those neurotoxins and summarize different neurotoxic animal models with the hope to help researchers choose among them accurately and to promote the development of modeling PD.
Collapse
Affiliation(s)
- Xian-Si Zeng
- College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| | - Wen-Shuo Geng
- College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| | - Jin-Jing Jia
- College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, China
| |
Collapse
|
124
|
Zhang J, Tong W, Sun H, Jiang M, Shen Y, Liu Y, Gu H, Guo J, Fang J, Jin L. Nrf2-mediated neuroprotection by MANF against 6-OHDA-induced cell damage via PI3K/AKT/GSK3β pathway. Exp Gerontol 2017; 100:77-86. [DOI: 10.1016/j.exger.2017.10.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/14/2017] [Accepted: 10/23/2017] [Indexed: 01/13/2023]
|
125
|
Neuroprotective and Neuro-restorative Effects of Minocycline and Rasagiline in a Zebrafish 6-Hydroxydopamine Model of Parkinson's Disease. Neuroscience 2017; 367:34-46. [PMID: 29079063 DOI: 10.1016/j.neuroscience.2017.10.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/15/2017] [Accepted: 10/16/2017] [Indexed: 02/01/2023]
Abstract
Parkinson's disease is a common, debilitating, neurodegenerative disorder for which the current gold standard treatment, levodopa (L-DOPA) is symptomatic. There is an urgent, unmet need for neuroprotective or, ideally, neuro-restorative drugs. We describe a 6-hydroxydopamine (6-OHDA) zebrafish model to screen drugs for neuroprotective and neuro-restorative capacity. Zebrafish larvae at two days post fertilization were exposed to 6-OHDA for three days, with co-administration of test drugs for neuroprotection experiments, or for 32 h, with subsequent treatment with test drugs for neuro-restoration experiments. Locomotor activity was assessed by automated tracking and dopaminergic neurons were visualized by tyrosine hydroxylase immuno-histochemistry. Exposure to 6-OHDA for either 32 h or 3 days induced similar, significant locomotor deficits and neuronal loss in 5-day-old larvae. L-DOPA (1 mM) partially restored locomotor activity, but was neither neuroprotective nor neuro-restorative, mirroring the clinical situation. The calcium channel blocker, isradipine (1 µM) did not prevent or reverse 6-OHDA-induced locomotor deficit or neuronal loss. However, both the tetracycline analog, minocycline (10 µM), and the monoamine oxidase B inhibitor, rasagiline (1 µM), prevented the locomotor deficits and neuronal loss due to three-day 6-OHDA exposure. Importantly, they also reversed the locomotor deficit caused by prior exposure to 6-OHDA; rasagiline also reversed neuronal loss and minocycline partially restored neuronal loss due to prior 6-OHDA, making them candidates for investigation as neuro-restorative treatments for Parkinson's disease. Our findings in zebrafish reflect preliminary clinical findings for rasagiline and minocycline. Thus, we have developed a zebrafish model suitable for high-throughput screening of putative neuroprotective and neuro-restorative therapies for the treatment of Parkinson's disease.
Collapse
|
126
|
Neurotrophic function of phytochemicals for neuroprotection in aging and neurodegenerative disorders: modulation of intracellular signaling and gene expression. J Neural Transm (Vienna) 2017; 124:1515-1527. [PMID: 29030688 DOI: 10.1007/s00702-017-1797-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/05/2017] [Indexed: 02/07/2023]
Abstract
Bioactive compounds in food and beverages have been reported to promote health and prevent age-associated decline in cognitive, motor and sensory activities, and emotional function. Phytochemicals, a ubiquitous class of plant secondary metabolites, protect neuronal cells by interaction with cellular activities, in addition to the antioxidant and anti-inflammatory function. In aging and age-associated neurodegenerative disorders, phytochemicals protect neuronal cells by neurotrophic factor-mimic activity, in addition to suppression of apoptosis signaling in mitochondria. This review presents the cellular mechanisms underlying anti-apoptotic function and neurotrophic function of phytochemicals in the brain. Phytochemicals bind to receptors of neurotrophic factors, and also receptors for γ-aminobutyric acid, acetylcholine, serotonin, and glutamate and estrogen, and activate downstream signal pathways. Phytochemicals also directly intervene intracellular signaling molecules to modify the brain function. Finally, phytochemicals enhance the endogenous biosynthesis of genes coding anti-apoptotic Bcl-2 and neurotrophic factors, such as brain-derived and glial cell line-derived neurotrophic factor. The gene induction may play a major role in the neuroprotective function of dietary compounds shown by epidemiological studies. Quantitative measurement of neurotrophic factors induced by phytochemicals in the serum, cerebrospinal fluid, and other clinical samples is proposed as a surrogate assay method to evaluate the neuroprotective potency. Development of novel neuroprotective compounds is expected among compounds chemically synthesized from the brain-permeable basic structure of phytochemicals.
Collapse
|
127
|
Neural Stem Cell Transplantation Promotes Functional Recovery from Traumatic Brain Injury via Brain Derived Neurotrophic Factor-Mediated Neuroplasticity. Mol Neurobiol 2017; 55:2696-2711. [PMID: 28421542 DOI: 10.1007/s12035-017-0551-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 04/07/2017] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) induces cognitive impairments, motor and behavioral deficits. Previous evidences have suggested that neural stem cell (NSC) transplantation could facilitate functional recovery from brain insults, but their underlying mechanisms remains to be elucidated. Here, we established TBI model by an electromagnetic-controlled cortical impact device in the rats. Then, 5 μl NSCs (5.0 × 105/μl), derived from green fluorescent protein (GFP) transgenic mouse, was transplanted into the traumatic brain regions of rats at 24 h after injury. After differentiation of the NSCs was determined using immunohistochemistry, neurological severity scores (NSS) and rotarod test were conducted to detect the neurological behavior. Western blot and RT-PCR as well as ELASA were used to evaluate the expression of synaptophysin and brain-derived neurotrophic factor (BDNF). In order to elucidate the role of BDNF on the neural recovery after NSC transplantation, BDNF knockdown in NSC was performed and transplanted into the rats with TBI, and potential mechanism for BDNF knockdown in the NSC was analyzed using microassay analysis. Meanwhile, BDNF antibody blockade was conducted to further confirm the effect of BDNF on neural activity. As a result, an increasing neurological function improvement was seen in NSC transplanted rats, which was associated with the upregulation of synaptophysin and BDNF expression. Moreover, transplantation of BDNF knockdown NSCs and BDNF antibody block reduced not only the level of synaptophysin but also exacerbated neurological function deficits. Microassay analysis showed that 14 genes such as Wnt and Gsk3-β were downregulated after BDNF knockdown. The present data therefore showed that BDNF-mediated neuroplasticity underlie the mechanism of NSC transplantation for the treatment of TBI in adult rats.
Collapse
|
128
|
Sun Z, Hu W, Yin S, Lu X, Zuo W, Ge S, Xu Y. NGF protects against oxygen and glucose deprivation-induced oxidative stress and apoptosis by up-regulation of HO-1 through MEK/ERK pathway. Neurosci Lett 2017; 641:8-14. [PMID: 28115238 DOI: 10.1016/j.neulet.2017.01.046] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/18/2017] [Accepted: 01/19/2017] [Indexed: 01/28/2023]
Abstract
Both nerve growth factor (NGF) and heme oxygenases-1 (HO-1) promotes neuron survival from cerebral ischemic lesions. NGF protects neurons from oxygen-glucose deprivation (OGD), and HO-1 expression can be induced by some growth factors like NGF. This work attempted to identify the contribution of HO-1 on the neuroprotection role of NGF in OGD model, which is an injury simulation of ischemic neuron in vitro. The viability of cortical neurons cells treated with OGD restored significantly by pretreatment with NGF in a dose dependent manner. Moreover, NGF provided obvious protective effects against OGD-induced neurons apoptosis. It identified that NGF could prevent apoptosis and ROS (reactive oxygen species) accumulation in the primary cortical neurons exposed to OGD. NGF could up-regulate the expression level of HO-1, and then afford neuroprotection against OGD insult. In addition, we found that MEK/ERK pathway participated NGF-induced over-expression of HO-1, and was involved in the transcriptional activity or neuroprotection effect of NGF.
Collapse
Affiliation(s)
- Zhitang Sun
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurology, the Second Hospital of Shanxi Medical University,382 Wuyi Road, Taiyuan 030001, Shanxi, China
| | - Weimin Hu
- Department of Neurology, the Second Hospital of Shanxi Medical University,382 Wuyi Road, Taiyuan 030001, Shanxi, China
| | - Shulan Yin
- Department of Neurology, the Second Hospital of Shanxi Medical University,382 Wuyi Road, Taiyuan 030001, Shanxi, China
| | - Xiufang Lu
- Department of Neurology, the Second Hospital of Shanxi Medical University,382 Wuyi Road, Taiyuan 030001, Shanxi, China
| | - Wenchao Zuo
- Department of Neurology, the Second Hospital of Shanxi Medical University,382 Wuyi Road, Taiyuan 030001, Shanxi, China
| | - Sihui Ge
- Department of Neurology, the Second Hospital of Shanxi Medical University,382 Wuyi Road, Taiyuan 030001, Shanxi, China
| | - Yuming Xu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
129
|
Yin P, Zhang J, Yan L, Yang L, Sun L, Shi L, Ma C, Liu Y. Urolithin C, a gut metabolite of ellagic acid, induces apoptosis in PC12 cells through a mitochondria-mediated pathway. RSC Adv 2017. [DOI: 10.1039/c7ra01548h] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Urolithin C includes apoptosis in PC12 cells through a mitochondria-mediated pathway.
Collapse
Affiliation(s)
- Peipei Yin
- National Engineering Laboratory for Tree Breeding
- College of Biological Sciences and Biotechnology
- Beijing Forestry University
- Beijing 100083
- China
| | - Jianwei Zhang
- National Engineering Laboratory for Tree Breeding
- College of Biological Sciences and Biotechnology
- Beijing Forestry University
- Beijing 100083
- China
| | - Linlin Yan
- National Engineering Laboratory for Tree Breeding
- College of Biological Sciences and Biotechnology
- Beijing Forestry University
- Beijing 100083
- China
| | - Lingguang Yang
- National Engineering Laboratory for Tree Breeding
- College of Biological Sciences and Biotechnology
- Beijing Forestry University
- Beijing 100083
- China
| | - Liwei Sun
- National Engineering Laboratory for Tree Breeding
- College of Biological Sciences and Biotechnology
- Beijing Forestry University
- Beijing 100083
- China
| | - Lingling Shi
- National Engineering Laboratory for Tree Breeding
- College of Biological Sciences and Biotechnology
- Beijing Forestry University
- Beijing 100083
- China
| | - Chao Ma
- National Engineering Laboratory for Tree Breeding
- College of Biological Sciences and Biotechnology
- Beijing Forestry University
- Beijing 100083
- China
| | - Yujun Liu
- National Engineering Laboratory for Tree Breeding
- College of Biological Sciences and Biotechnology
- Beijing Forestry University
- Beijing 100083
- China
| |
Collapse
|