1
|
Seo CR, Lee BK, Jee HJ, Yoo JR, Lee CK, Park JW, Jung YS. Ameliorating Effect of Fermented Perilla frutescens on Sleep Deprivation-Induced Cognitive Impairment Through Antioxidant and BDNF Signaling in Mice. Nutrients 2024; 16:4224. [PMID: 39683616 DOI: 10.3390/nu16234224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Adequate sleep is essential for maintaining cognitive function, as evidenced by literature. Perilla frutescens var. acuta Kudo (PF) is a traditional medicinal herb reported to improve vascular cognitive impairment and induce sedation. However, the effects of PF on cognitive impairment caused by sleep deprivation (SD) have not yet been evaluated. This study aims to evaluate the effects of fermented PF (FPF) and its underlying mechanisms in a model of SD-induced cognitive impairment. Methods: Mice were subjected to SD to establish cognitive impairment, and FPF was administered once daily for 3 days. Cognitive performance was assessed using Y-maze and passive avoidance tests, followed by molecular mechanisms analyses. Results: FPF treatment improved SD-induced cognitive impairment, as evidenced by increased spontaneous alternation and extended latency time. Histological analysis revealed that SD impaired the hippocampus, and this impairment was alleviated by FPF treatment. FPF demonstrated antioxidant activity by increasing glutathione levels and decreasing malondialdehyde levels. Furthermore, the decreased levels of brain-derived neurotrophic factor (BDNF) observed in sleep-deprived mice were restored with FPF treatment. FPF also enhanced the phosphorylation of tropomyosin receptor kinase B, extracellular signal-regulated kinase, and cAMP response element-binding protein. Conclusions: These results indicate that FPF may have beneficial effects on SD-induced cognitive impairment by protecting against oxidative stress and increasing BDNF expression.
Collapse
Affiliation(s)
- Chae-Ryeong Seo
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Bo Kyung Lee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Hye Jin Jee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Jae Ryeong Yoo
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Chul-Kyu Lee
- Headquarters of New Drug Development Support, Corestemchemon Inc., 15 F, Gyeonggi Bio Center, Suwon 16229, Republic of Korea
| | - Jin Wook Park
- Headquarters of New Drug Development Support, Corestemchemon Inc., 15 F, Gyeonggi Bio Center, Suwon 16229, Republic of Korea
| | - Yi-Sook Jung
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
2
|
Park SY, Kim KY, Gwak DS, Shin SY, Jun DY, Kim YH. L-Cysteine mitigates ROS-induced apoptosis and neurocognitive deficits by protecting against endoplasmic reticulum stress and mitochondrial dysfunction in mouse neuronal cells. Biomed Pharmacother 2024; 180:117538. [PMID: 39393330 DOI: 10.1016/j.biopha.2024.117538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
Oxidative stress and mitochondrial dysfunction play critical roles in neurodegenerative diseases. Glutathione (GSH), a key brain antioxidant, helps to neutralize reactive oxygen species (ROS) and maintain redox balance. We investigated the effectiveness of L-cysteine (L-Cys) in preventing apoptosis induced by the ROS generator 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) in mouse hippocampal neuronal HT22 cells, as well as alleviating memory and cognitive impairments caused by the GSH synthesis inhibitor L-buthionine sulfoximine (BSO) in mice. DMNQ-induced apoptotic events in HT22 cells, including elevated cytosolic and mitochondrial ROS levels, DNA fragmentation, endoplasmic reticulum stress, and mitochondrial damage-mediated apoptotic pathways were dose-dependently abrogated by L-Cys (0.5-2 mM). The reduced intracellular GSH level, caused by DMNQ treatment, was restored by L-Cys cotreatment. Although L-Cys did not significantly restore GSH in the presence of BSO, it prevented DMNQ-induced ROS elevation, mitochondrial damage, and apoptosis. Furthermore, compared to N-acetylcysteine and GSH, L-Cys had higher 2,2-diphenyl-1-picrylhydrazyl and 2,2-azino-bis-3-ethylbenzothiazoline-6-sulphonic acid radical-scavenging activity. L-Cys also restored mitochondrial respiration capacity in DMNQ-treated HT22 cells by reversing mitochondrial fission-fusion dynamic balance. BSO administration (500 mg/kg/day) in mice led to neuronal deficits, including memory and cognitive impairments, which were effectively mitigated by oral L-Cys (15 or 30 mg/kg/day). L-Cys also reduced BSO-induced ROS levels in the mice hippocampus and cortex. These findings suggest that even though it does not contribute to intracellular GSH synthesis, exogenous L-Cys protects neuronal cells against oxidative stress-induced mitochondrial damage and apoptosis, by acting as a ROS scavenger, which is beneficial in ameliorating neurocognitive deficits caused by oxidative stress.
Collapse
Affiliation(s)
- Shin Young Park
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Ki Yun Kim
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Dong Seol Gwak
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Do Youn Jun
- AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Young Ho Kim
- Laboratory of Immunobiology, School of Life Science, College of Natural Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; AT-31 BIO Inc., Business Incubation Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea.
| |
Collapse
|
3
|
Romanowski G, Budka J, Inkielewicz-Stepniak I. Oxidovanadium(V) Schiff Base Complexes Derived from Chiral 3-amino-1,2-propanediol Enantiomers: Synthesis, Spectroscopic Studies, Catalytic and Biological Activity. Int J Mol Sci 2024; 25:5010. [PMID: 38732229 PMCID: PMC11084397 DOI: 10.3390/ijms25095010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Oxidovanadium(V) complexes, [(+)VOL1-5] and [(-)VOL1-5], with chiral tetradentate Schiff bases, which are products of monocondensation of S(‒)-3-amino-1,2-propanediol or R(+)-3-amino-1,2-propanediol with salicylaldehyde derivatives, have been synthesized. Different spectroscopic methods, viz. 1H and 51V NMR, IR, UV-Vis, and circular dichroism, as well as elemental analysis, have been used for their detailed characterization. Furthermore, the epoxidation of styrene, cyclohexene, and two monoterpenes, S(‒)-limonene and (‒)-α-pinene, using two oxidants, aqueous 30% H2O2 or tert-butyl hydroperoxide (TBHP) in decane, has been studied with catalytic amounts of all complexes. Finally, biological cytotoxicity studies have also been performed with these oxidovanadium(V) compounds for comparison with cis-dioxidomolybdenum(VI) Schiff base complexes with the same chiral ligands, as well as to determine the cytoprotection against the oxidative damage caused by 30% H2O2 in the HT-22 hippocampal neuronal cells in the range of their 10-100 μM concentration.
Collapse
Affiliation(s)
- Grzegorz Romanowski
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, PL-80308 Gdansk, Poland
| | - Justyna Budka
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Dębinki 7, Building 27, PL-80211 Gdansk, Poland;
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Dębinki 7, Building 27, PL-80211 Gdansk, Poland;
| |
Collapse
|
4
|
Goujon M, Liang Z, Soriano-Castell D, Currais A, Maher P. The Neuroprotective Flavonoids Sterubin and Fisetin Maintain Mitochondrial Health under Oxytotic/Ferroptotic Stress and Improve Bioenergetic Efficiency in HT22 Neuronal Cells. Antioxidants (Basel) 2024; 13:460. [PMID: 38671908 PMCID: PMC11047672 DOI: 10.3390/antiox13040460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The global increase in the aging population has led to a rise in many age-related diseases with continuing unmet therapeutic needs. Research into the molecular mechanisms underlying both aging and neurodegeneration has identified promising therapeutic targets, such as the oxytosis/ferroptosis cell death pathway, in which mitochondrial dysfunction plays a critical role. This study focused on sterubin and fisetin, two flavonoids from the natural pharmacopeia previously identified as strong inhibitors of the oxytosis/ferroptosis pathway. Here, we investigated the effects of the compounds on the mitochondrial physiology in HT22 hippocampal nerve cells under oxytotic/ferroptotic stress. We show that the compounds can restore mitochondrial homeostasis at the level of redox regulation, calcium uptake, biogenesis, fusion/fission dynamics, and modulation of respiration, leading to the enhancement of bioenergetic efficiency. However, mitochondria are not required for the neuroprotective effects of sterubin and fisetin, highlighting their diverse homeostatic impacts. Sterubin and fisetin, thus, provide opportunities to expand drug development strategies for anti-oxytotic/ferroptotic agents and offer new perspectives on the intricate interplay between mitochondrial function, cellular stress, and the pathophysiology of aging and age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Marie Goujon
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| | - Zhibin Liang
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| | - David Soriano-Castell
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| | - Antonio Currais
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| |
Collapse
|
5
|
Shi J, Li W, Ding X, Zhou F, Hao C, He M, Wang F, Li X. The role of the SIRT1-BMAL1 pathway in regulating oxidative stress in the early development of ischaemic stroke. Sci Rep 2024; 14:1773. [PMID: 38245621 PMCID: PMC10799848 DOI: 10.1038/s41598-024-52120-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/14/2024] [Indexed: 01/22/2024] Open
Abstract
Oxidative stress is the primary cause of ischaemic stroke and is closely related to circadian rhythm. However, the mechanism by which circadian rhythm regulates oxidative stress in ischaemic stroke remains elusive. The Silent Information Regulator 1 (SIRT1) controls circadian rhythm by activating the transcription of the circadian clock core protein Basic Helix-Loop-Helix ARNT Like 1 (BMAL1) through deacetylation. Studies have shown that the SIRT1-BMAL1 pathway can regulate oxidative stress. To investigate its correlation with oxidative stress, we examined the expression levels and influencing factors of SIRT1-BMAL1 at different times in ischaemic stroke patients and analyzed their clinical indexes, oxidative stress, and inflammatory factor indicators. The expression levels of oxidative stress and inflammatory factor indicators, including malondialdehyde (MDA), superoxide dismutase (SOD), interleukin-6 (IL-6), and tumor necrosis factor-a (TNF-α), SIRT1, and BMAL1, were detected in ischaemic stroke patients within 4.5 h of onset and in non-stroke patients. Patients were divided into four subgroups based on onset time: subgroup 1 (0:00-05:59); subgroup 2 (06:00-11:59); subgroup 3 (12:00-17: 59); and subgroup 4 (18:00-23:59). Our results showed higher MDA, IL-6, and TNF-α levels, and lower SOD, SIRT1, and BMAL1 levels in ischaemic stroke patients compared to control patients (P < 0.05). Among the four subgroups, the content of MDA, IL-6, and TNF-α was highest in patients with ischaemic stroke onset from subgroup 2 (06:00-11:59), while the expression levels of SOD, BMAL1, and SIRT1 were lowest in patients with ischaemic stroke in subgroup 2. Additionally, myeloperoxidase (MPO) reached the highest value showing the same trends consistent with MDA, IL-6, and TNF-ɑ and opposite trends consistent with SOD, BMAL1, and SIRT1. However, triglycerides (TGs), total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), immediate blood glucose, immediate diastolic blood pressure, immediate systolic blood pressure, and homocysteine (HCY) did not show any statistically significant circadian rhythm changes (P > 0.05). Our findings suggest that the SIRT1-BMAL1 pathway may be involved in early oxidative stress in ischaemic stroke, which may be related to MPO.
Collapse
Affiliation(s)
- Jing Shi
- Department of Neurology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
- The Ninth Clinical Medical College Affiliated with Shanxi Medical University, Taiyuan, China
| | - Weirong Li
- Cardiovascular Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xiaobo Ding
- School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Feng Zhou
- The Ninth Clinical Medical College Affiliated with Shanxi Medical University, Taiyuan, China
| | - Chenxi Hao
- The First Clinical Medical College Affiliated with Shanxi Medical University, Taiyuan, China
| | - Miao He
- The Ninth Clinical Medical College Affiliated with Shanxi Medical University, Taiyuan, China
| | - Fan Wang
- Department of Neurology, Aerospace Center Hospital, Peking University Aerospace Clinic College of Medicine, Beijing, China.
| | - Xinyi Li
- Department of Neurology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China.
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Romanowski G, Budka J, Inkielewicz-Stepniak I. Synthesis, Spectroscopic Characterization, Catalytic and Biological Activity of Oxidovanadium(V) Complexes with Chiral Tetradentate Schiff Bases. Molecules 2023; 28:7408. [PMID: 37959827 PMCID: PMC10649191 DOI: 10.3390/molecules28217408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
New oxidovanadium(V) complexes, VOL1-VOL10, with chiral tetradentate Schiff bases obtained by monocondensation reaction of salicylaldehyde derivatives with 1S,2S-(+)-2-amino-1-(4-nitrophenyl)-1,3-propanediol. All complexes have been characterized using different spectroscopic methods, viz. IR, UV-Vis, circular dichroism, one- (1H, 51V) and two-dimensional (COSY, NOESY) NMR spectroscopy, and elemental analysis. Furthermore, the catalytic ability of all compounds in the epoxidation of styrene, cyclohexene, and its naturally occurring monoterpene derivatives, i.e., S(-)-limonene and (-)-α-pinene has also been studied, using two different oxidants, i.e., aqueous 30% H2O2 or tert-butyl hydroperoxide (TBHP). In addition, the biological properties of these chiral oxidovanadium(V) compounds, but also cis-dioxidomolybdenum(VI) complexes with the same chiral Schiff bases, were studied. Their cytotoxic and cytoprotective activity studies with the HT-22 hippocampal neuronal cells revealed a concentration-dependent effect in the range of 10-100 μM. Moreover, vanadium(V) complexes, in contrast to cis-dioxidomolybdenum(VI) compounds, demonstrated higher cytotoxicity and lack of cytoprotective ability against H2O2-induced cytotoxicity.
Collapse
Affiliation(s)
- Grzegorz Romanowski
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, PL-80308 Gdansk, Poland
| | - Justyna Budka
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Dębinki 7, Building 27, PL-80211 Gdansk, Poland
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Dębinki 7, Building 27, PL-80211 Gdansk, Poland
| |
Collapse
|
7
|
Pham TNM, Perumal N, Manicam C, Basoglu M, Eimer S, Fuhrmann DC, Pietrzik CU, Clement AM, Körschgen H, Schepers J, Behl C. Adaptive responses of neuronal cells to chronic endoplasmic reticulum (ER) stress. Redox Biol 2023; 67:102943. [PMID: 37883843 PMCID: PMC10618786 DOI: 10.1016/j.redox.2023.102943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Accumulation of misfolded proteins or perturbation of calcium homeostasis leads to endoplasmic reticulum (ER) stress and is linked to the pathogenesis of neurodegenerative diseases. Hence, understanding the ability of neuronal cells to cope with chronic ER stress is of fundamental interest. Interestingly, several brain areas uphold functions that enable them to resist challenges associated with neurodegeneration. Here, we established novel clonal mouse hippocampal (HT22) cell lines that are resistant to prolonged (chronic) ER stress induced by thapsigargin (TgR) or tunicamycin (TmR) as in vitro models to study the adaption to ER stress. Morphologically, we observed a significant increase in vesicular und autophagosomal structures in both resistant lines and 'giant lysosomes', especially striking in TgR cells. While autophagic activity increased under ER stress, lysosomal function appeared slightly impaired; in both cell lines, we observed enhanced ER-phagy. However, proteomic analyses revealed that various protein clusters and signaling pathways were differentially regulated in TgR versus TmR cells in response to chronic ER stress. Additionally, bioenergetic analyses in both resistant cell lines showed a shift toward aerobic glycolysis ('Warburg effect') and a defective complex I of the oxidative phosphorylation (OXPHOS) machinery. Furthermore, ER stress-resistant cells differentially activated the unfolded protein response (UPR) comprising IRE1α and ATF6 pathways. These findings display the wide portfolio of adaptive responses of neuronal cells to chronic ER stress. ER stress-resistant neuronal cells could be the basis to uncover molecular modulators of adaptation, resistance, and neuroprotection as potential pharmacological targets for preventing neurodegeneration.
Collapse
Affiliation(s)
- Thu Nguyen Minh Pham
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Natarajan Perumal
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Marion Basoglu
- Department of Structural Cell Biology, Institute for Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
| | - Stefan Eimer
- Department of Structural Cell Biology, Institute for Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Albrecht M Clement
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hagen Körschgen
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jana Schepers
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Christian Behl
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
8
|
Yin Z, You S, Zhang S, Zhang L, Wu B, Huang X, Lu S, Cao L, Zhang Y, Li D, Zhang X, Liu J, Sun Y, Zhang N. Atorvastatin rescues vascular endothelial injury in hypertension by WWP2-mediated ubiquitination and degradation of ATP5A. Biomed Pharmacother 2023; 166:115228. [PMID: 37557013 DOI: 10.1016/j.biopha.2023.115228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023] Open
Abstract
As a widely used lipid-lowering drug in clinical practice, atorvastatin is widely recognized for its role in protecting vascular endothelium in the cardiovascular system. However, a clear mechanistic understanding of its action is lacking. Here, we found that atorvastatin counteracted angiotensin II-induced vascular endothelial injury in mice with hypertension. Mechanistically, atorvastatin up-regulated WWP2, a E6AP C-terminus (HECT)-type E3 ubiquitin ligase with an essential role in regulating protein ubiquitination and various biological processes, thereby rescuing vascular endothelial injury. By ubiquitinating ATP5A (ATP synthase mitochondrial F1 complex subunit alpha), WWP2 degraded ATP5A via the proteasome pathway, stabilizing Bcl-2/Bax in the mitochondrial pathway of apoptosis. Moreover, atorvastatin further ameliorated death of vascular endothelial cells and improved vascular endothelial functions under WWP2 overexpression, whereas WWP2 knockout abrogated these beneficial effects of atorvastatin. Furthermore, we generated endothelial cell-specific WWP2 knockout mice, and this WWP2-mediated mechanism was faithfully recapitulated in vivo. Thus, we propose that activation of a WWP2-dependent pathway that is pathologically repressed in damaged vascular endothelium under hypertension is a major mechanism of atorvastatin. Our findings are also pertinent to develop novel therapeutic strategies for vascular endothelial injury-related cardiovascular diseases.
Collapse
Affiliation(s)
- Zeyu Yin
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shu Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Linlin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinyue Huang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Saien Lu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Da Li
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, Liaoning Province, China; Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xingang Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jingwei Liu
- Department of Anus and Intestine Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, Liaoning Province, China.
| |
Collapse
|
9
|
Konadu B, Hosler JP, Gibert Y, Edwards KS. Analysis of oxygen consumption rates in zebrafish reveals differences based on sex, age and physical activity recovery. Front Physiol 2023; 14:1272366. [PMID: 37781232 PMCID: PMC10534073 DOI: 10.3389/fphys.2023.1272366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction: Mitochondrial dysfunction is linked to a variety of human diseases. Understanding the dynamic alterations in mitochondrial respiration at various stages of development is important to our understanding of disease progression. Zebrafish provide a system for investigating mitochondrial function and alterations during different life stages. The purpose of this study was to investigate our ability to measure mitochondrial oxygen consumption rates in zebrafish embryos, larvae, and adults as an indicator of mitochondrial function. Methods: Basal respiration of entire zebrafish embryos (5 dpf), larvae (0.6-0.9 cm), young adults (3-month-old), and old adults (12-month-old) was measured using an Oroboros Oxygraph, with a stirrer speed of 26 rpm. For embryos and larvae, "leak" respiration (plus oligomycin), maximum respiration (plus uncoupler), non-mitochondrial respiration (plus inhibitors), and complex IV activity were also measured. To induce physical activity in adult fish, the stirrer speed was increased to 200 rpm. Results and Discussion: We demonstrate the ability to accurately measure respiration rates in zebrafish at various ages using the Oroboros Oxygraph. When comparing zebrafish embryos to larvae, embryos have a higher maximum respiration. Three-month-old zebrafish males have higher basal respiration than females, while 12-month-old zebrafish females exhibit greater rates of respiration than males and younger females. When the stirrer speed was increased, respiration rates decrease, but with differences depending on sex. This study demonstrates a simple and accessible method to assess zebrafish physiology by mitochondrial oxygen consumption measurements in an unmodified Oroboros Oxygraph. The method should facilitate studies to understand the intricate interplay between mitochondrial function, development, and aging.
Collapse
Affiliation(s)
- Bridget Konadu
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jonathan P. Hosler
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yann Gibert
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, United States
| | - Kristin S. Edwards
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
10
|
Endres K, Friedland K. Talk to Me-Interplay between Mitochondria and Microbiota in Aging. Int J Mol Sci 2023; 24:10818. [PMID: 37445995 DOI: 10.3390/ijms241310818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The existence of mitochondria in eukaryotic host cells as a remnant of former microbial organisms has been widely accepted, as has their fundamental role in several diseases and physiological aging. In recent years, it has become clear that the health, aging, and life span of multicellular hosts are also highly dependent on the still-residing microbiota, e.g., those within the intestinal system. Due to the common evolutionary origin of mitochondria and these microbial commensals, it is intriguing to investigate if there might be a crosstalk based on preserved common properties. In the light of rising knowledge on the gut-brain axis, such crosstalk might severely affect brain homeostasis in aging, as neuronal tissue has a high energy demand and low tolerance for according functional decline. In this review, we summarize what is known about the impact of both mitochondria and the microbiome on the host's aging process and what is known about the aging of both entities. For a long time, bacteria were assumed to be immortal; however, recent evidence indicates their aging and similar observations have been made for mitochondria. Finally, we present pathways by which mitochondria are affected by microbiota and give information about therapeutic anti-aging approaches that are based on current knowledge.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Kristina Friedland
- Department of Pharmacology and Toxicology, Institute for Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, 55128 Mainz, Germany
| |
Collapse
|
11
|
Thiamine insufficiency induces Hypoxia Inducible Factor-1α as an upstream mediator for neurotoxicity and AD-like pathology. Mol Cell Neurosci 2022; 123:103785. [PMID: 36241022 DOI: 10.1016/j.mcn.2022.103785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022] Open
Abstract
Insufficiencies of the micronutrient thiamine (Vitamin B1) have been associated with inducing Alzheimer's disease (AD)-like neuropathology. The hypometabolic state associated with chronic thiamine insufficiency (TI) has been demonstrated to be a contributor towards the development of amyloid plaque deposition and neurotoxicity. However, the molecular mechanism underlying TI induced AD pathology is still unresolved. Previously, we have established that TI stabilizes the metabolic stress transcriptional factor, Hypoxia Inducible Factor-1α (HIF1α). Utilizing neuronal hippocampal cells (HT22), TI-induced HIF1α activation triggered the amyloidogenic cascade through transcriptional expression and increased activity of β-secretase (BACE1). Knockdown and pharmacological inhibition of HIF1α during TI significantly reduced BACE1 and C-terminal Fragment of 99 amino acids (C99) formation. TI also increased the expression of the HIF1α regulated pro-apoptotic protein, BCL2/adenovirus E1B 19 kDa protein-interacting protein (BNIP3). Correspondingly, cell toxicity during TI conditions was significantly reduced with HIF1α and BNIP3 knockdown. The role of BNIP3 in TI-mediated toxicity was further highlighted by localization of dimeric BNIP3 into the mitochondria and nuclear accumulation of Endonuclease G. Subsequently, TI decreased mitochondrial membrane potential and enhanced chromatin fragmentation. However, cell toxicity via the HIF1α/BNIP3 cascade required TI induced oxidative stress. HIF1α, BACE1 and BNIP3 expression was induced in 3xTg-AD mice after TI and administration with the HIF1α inhibitor YC1 significantly attenuated HIF1α and target genes levels in vivo. Overall, these findings demonstrate a critical stress response during TI involving the induction of HIF1α transcriptional activity that directly promotes neurotoxicity and AD-like pathology.
Collapse
|
12
|
Jeon KB, Lee SH, Kwon YS, Beak JH, Lee H, Ma CJ. Protective effect of fermented aloe extract on glutamate-induced cytotoxicity in HT22 cells. Anim Cells Syst (Seoul) 2022; 26:318-327. [PMID: 36605589 PMCID: PMC9809408 DOI: 10.1080/19768354.2022.2147584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Excessive glutamate can cause oxidative stress in neuronal cells and this can significantly contribute to the etiology of neurodegenerative disease. The present study mainly aims to investigate that aloe extract (AE) and fermented aloe extract (FAE) could protect against glutamate-induced cytotoxicity by modulating oxidative stress. In this study, both AE and FAE showed potent neuroprotective activity by inhibiting ROS and Ca2+ concentration, increasing mitochondria membrane potential, and activating glutathione-related enzymes against glutamate-insulted neurotoxicity in HT22 cells. In addition, the neuroprotective activity of FAE was more potent than that of AE. HPLC analysis reveals that the chemical composition of FAE is different from that of AE. Especially, the contents of aloin A, aloin B and aloenin were higher in FAE than in AE. In conclusion, this study indicates that both AE and FAE may have effective neuroprotective activity in glutamate-insulted pathological conditions such as Alzheimer's disease by managing oxidative stress.
Collapse
Affiliation(s)
- Ki Beom Jeon
- R&DB Center, Beauty Science, Ltd., Sejong, Korea
| | | | | | | | - Hyeon Lee
- R&DB Center, Beauty Science, Ltd., Sejong, Korea
| | - Choong Je Ma
- Department of Medical Biomaterials Engineering, College of Biomedical Science, Kangwon National University, Chuncheon, South Korea,Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, South Korea, Choong Je Department of Medical Biomaterials Engineering, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, South Korea. Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, 24341, South Korea.
| |
Collapse
|
13
|
van der Kooij MA, Rojas-Charry L, Givehchi M, Wolf C, Bueno D, Arndt S, Tenzer S, Mattioni L, Treccani G, Hasch A, Schmeisser MJ, Vianello C, Giacomello M, Methner A. Chronic social stress disrupts the intracellular redistribution of brain hexokinase 3 induced by shifts in peripheral glucose levels. J Mol Med (Berl) 2022; 100:1441-1453. [PMID: 35943566 PMCID: PMC9470722 DOI: 10.1007/s00109-022-02235-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/06/2022] [Accepted: 07/04/2022] [Indexed: 11/26/2022]
Abstract
Abstract
Chronic stress has the potential to impair health and may increase the vulnerability for psychiatric disorders. Emerging evidence suggests that specific neurometabolic dysfunctions play a role herein. In mice, chronic social defeat (CSD) stress reduces cerebral glucose uptake despite hyperglycemia. We hypothesized that this metabolic decoupling would be reflected by changes in contact sites between mitochondria and the endoplasmic reticulum, important intracellular nutrient sensors, and signaling hubs. We thus analyzed the proteome of their biochemical counterparts, mitochondria-associated membranes (MAMs) from whole brain tissue obtained from CSD and control mice. This revealed a lack of the glucose-metabolizing enzyme hexokinase 3 (HK3) in MAMs from CSD mice. In controls, HK3 protein abundance in MAMs and also in striatal synaptosomes correlated positively with peripheral blood glucose levels, but this connection was lost in CSD. We conclude that the ability of HK3 to traffic to sites of need, such as MAMs or synapses, is abolished upon CSD and surmise that this contributes to a cellular dysfunction instigated by chronic stress.
Key messages Chronic social defeat (CSD) alters brain glucose metabolism CSD depletes hexokinase 3 (HK3) from mitochondria-associated membranes (MAMs) CSD results in loss of positive correlation between blood glucose and HK3 in MAMs and synaptosomes
Collapse
Affiliation(s)
| | - Liliana Rojas-Charry
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, Mainz, 55131, Germany.,Institute of Anatomy, Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Maryam Givehchi
- Leibniz Institute for Resilience Research (LIR), Mainz, 55122, Germany
| | - Christina Wolf
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Diones Bueno
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Sabine Arndt
- Institute for Immunology, Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Stefan Tenzer
- Institute for Immunology, Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Lorenzo Mattioni
- Institute of Anatomy, Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Giulia Treccani
- Institute of Anatomy, Johannes Gutenberg University Mainz, Mainz, 55131, Germany.,Department of Psychiatry and Psychotherapy, Translational Psychiatry, University Medical Center, Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Annika Hasch
- Leibniz Institute for Resilience Research (LIR), Mainz, 55122, Germany
| | - Michael J Schmeisser
- Institute of Anatomy, Johannes Gutenberg University Mainz, Mainz, 55131, Germany
| | - Caterina Vianello
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, Mainz, 55131, Germany.,Department of Biology, University of Padua, Padua, 35121, Italy
| | | | - Axel Methner
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, Mainz, 55131, Germany.
| |
Collapse
|
14
|
Liang Z, Soriano-Castell D, Kepchia D, Duggan BM, Currais A, Schubert D, Maher P. Cannabinol inhibits oxytosis/ferroptosis by directly targeting mitochondria independently of cannabinoid receptors. Free Radic Biol Med 2022; 180:33-51. [PMID: 34999187 PMCID: PMC8840979 DOI: 10.1016/j.freeradbiomed.2022.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/08/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022]
Abstract
The oxytosis/ferroptosis regulated cell death pathway recapitulates many features of mitochondrial dysfunction associated with the aging brain and has emerged as a potential key mediator of neurodegeneration. It has thus been proposed that the oxytosis/ferroptosis pathway can be used to identify novel drug candidates for the treatment of age-associated neurodegenerative diseases that act by preserving mitochondrial function. Previously, we identified cannabinol (CBN) as a potent neuroprotector. Here, we demonstrate that not only does CBN protect nerve cells from oxytosis/ferroptosis in a manner that is dependent on mitochondria and it does so independently of cannabinoid receptors. Specifically, CBN directly targets mitochondria and preserves key mitochondrial functions including redox regulation, calcium uptake, membrane potential, bioenergetics, biogenesis, and modulation of fusion/fission dynamics that are disrupted following induction of oxytosis/ferroptosis. These protective effects of CBN are at least partly mediated by the promotion of endogenous antioxidant defenses and the activation of AMP-activated protein kinase (AMPK) signaling. Together, our data highlight the potential of mitochondrially-targeted compounds such as CBN as novel oxytotic/ferroptotic inhibitors to rescue mitochondrial dysfunction as well as opportunities for the discovery and development of future neurotherapeutics.
Collapse
Affiliation(s)
- Zhibin Liang
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, United States; The Paul F. Glenn Center for Biology of Aging Research, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, United States.
| | - David Soriano-Castell
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - Devin Kepchia
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - Brendan M Duggan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, United States
| | - Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - David Schubert
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, United States; The Paul F. Glenn Center for Biology of Aging Research, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, United States.
| |
Collapse
|
15
|
Mitochondrial abnormalities in neurodegenerative models and possible interventions: Focus on Alzheimer's disease, Parkinson's disease, Huntington's disease. Mitochondrion 2020; 55:14-47. [PMID: 32828969 DOI: 10.1016/j.mito.2020.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/22/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022]
Abstract
Mitochondrial abnormalities in the brain are considered early pathological changes in neurogenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). The mitochondrial dysfunction in the brain can be induced by toxic proteins, including amyloid-beta (Aβ), phosphorylated tau, alpha-synuclein (α-syn) and mutant huntingtin (mtHTT). These proteins cause mitochondrial genome damage, increased oxidative stress, decreased mitochondrial membrane permeability, and diminished ATP production. Consequently, synaptic dysfunction, synaptic loss, neuronal apoptosis, and ultimately cognitive impairment are exhibited. Therefore, the restoration of mitochondrial abnormalities in the brain is an alternative intervention to delay the progression of neurodegenerative diseases in addition to reducing the level of toxic proteins, especially Aβ, and restored synaptic dysfunction by interventions. Here we comprehensively review mitochondrial alterations in the brain of neurodegenerative models, specifically AD, PD and HD, from both in vitro and in vivo studies. Additionally, the correlation between mitochondrial changes, cognitive function, and disease progression from in vivo studies is described. This review also summarizes interventions that possibly attenuate mitochondrial abnormalities in AD, PD and HD models from both in vitro and in vivo studies. This may lead to the introduction of novel therapies that target on brain mitochondria to delay the progression of AD, PD and HD.
Collapse
|
16
|
Bueno DC, Canto RFS, de Souza V, Andreguetti RR, Barbosa FAR, Naime AA, Dey PN, Wüllner V, Lopes MW, Braga AL, Methner A, Farina M. New Probucol Analogues Inhibit Ferroptosis, Improve Mitochondrial Parameters, and Induce Glutathione Peroxidase in HT22 Cells. Mol Neurobiol 2020; 57:3273-3290. [DOI: 10.1007/s12035-020-01956-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
|
17
|
Neuroprotective Activity of Methanolic Extract of Lysimachia christinae against Glutamate Toxicity in HT22 Cell and Its Protective Mechanisms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5352034. [PMID: 32419811 PMCID: PMC7201513 DOI: 10.1155/2020/5352034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 11/21/2022]
Abstract
Purpose Excessive glutamate amount can give oxidative stress to neuronal cells, and the accumulation of cell death can trigger the neurodegenerative disorders. In this study, we discovered the neuroprotective effect of Lysimachia christinae Hance in the mouse hippocampal HT22 cell line. Method Overnight incubated HT22 cells were pretreated with L. christinae extract dose dependently (1, 10, and 100 μg/ml). Followed by then, glutamate was treated. These treated cells were incubated several times again, and cell viability, accumulation of reactive oxygen species (ROS) and Ca2+, mitochondrial membrane potential (MMP), and glutathione-related enzyme amount were measured. Results As a result, L. christinae increases the cell viability by inhibiting the ROS and Ca2+ formation, recovering the level of MMP and enhancing the activity of glutathione production compared with only vehicle-treated groups. Conclusion These draw that L. christinae may remarkably decelerate the neurodegeneration by minimizing neuronal cell damage via oxidative stress.
Collapse
|
18
|
Abi Nahed R, Reynaud D, Lemaitre N, Lartigue S, Roelants C, Vaiman D, Benharouga M, Cochet C, Filhol O, Alfaidy N. Protein kinase CK2 contributes to placental development: physiological and pathological implications. J Mol Med (Berl) 2019; 98:123-133. [PMID: 31832700 DOI: 10.1007/s00109-019-01855-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/21/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022]
Abstract
Preeclampsia (PE) is the most threatening pathology of human pregnancy. Its development is thought to be due to a failure in the invasion of trophoblast cells that establish the feto-maternal circulation. Protein kinase CK2 is a ubiquitous enzyme reported to be involved in the control of cell invasion. CK2 consists of two subunits, a catalytic subunit, CK2α, and a regulatory subunit, CK2β. To date, no data exist regarding the expression and role of this enzyme in normal and PE pregnancies. We performed studies, at the clinical level using distinctive cohorts from early pregnancy (n = 24) and from PE (n = 23) and age-matched controls (n = 28); in vitro, using trophoblast cell lines; ex vivo, using placental explants; and in vivo, using PE mouse models. We demonstrated that (i) CK2 is more expressed during the late first trimester of pregnancy and is mainly localized in differentiated trophoblast cells, (ii) the inhibition of its enzymatic activity decreased the proliferation, migration, invasion, and syncytialization of trophoblast cells, both in 2D and 3D culture systems, and (iii) CK2 activity and the CK2α/CK2β protein ratio were increased in PE human placentas. The pattern and profile of CK2 expression were confirmed in gravid mice along with an increase in the PE mouse models. Altogether, our results demonstrate that CK2 plays an essential role in the establishment of the feto-maternal circulation and that its deregulation is associated with PE development. The increase in CK2 activity in PE might constitute a compensatory mechanism to ensure proper pregnancy progress.
Collapse
Affiliation(s)
- Roland Abi Nahed
- Institut National de la Santé et de la Recherche Médicale, Unité, 1036, Grenoble, France. .,Université Grenoble-Alpes, 38000, Grenoble, France. .,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France.
| | - Deborah Reynaud
- Institut National de la Santé et de la Recherche Médicale, Unité, 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France
| | - Nicolas Lemaitre
- Institut National de la Santé et de la Recherche Médicale, Unité, 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France
| | - Solene Lartigue
- Institut National de la Santé et de la Recherche Médicale, Unité, 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France
| | - Caroline Roelants
- Institut National de la Santé et de la Recherche Médicale, Unité, 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France.,Inovarion, Paris, France
| | - Daniel Vaiman
- Genomics, Epigenetics and Physiopathology of Reproduction, Institut Cochin, U1016 Inserm- UMR 8104 CNRS - Paris-Descartes University, Paris, France
| | - Mohamed Benharouga
- Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France.,Laboratoire de Chimie et Biologie des Métaux, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Grenoble, France
| | - Claude Cochet
- Institut National de la Santé et de la Recherche Médicale, Unité, 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France
| | - Odile Filhol
- Institut National de la Santé et de la Recherche Médicale, Unité, 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale, Unité, 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Interdisciplinary Research Institure of Grenoble (IRIG), Grenoble, France
| |
Collapse
|
19
|
Wolf C, Zimmermann R, Thaher O, Bueno D, Wüllner V, Schäfer MKE, Albrecht P, Methner A. The Charcot-Marie Tooth Disease Mutation R94Q in MFN2 Decreases ATP Production but Increases Mitochondrial Respiration under Conditions of Mild Oxidative Stress. Cells 2019; 8:cells8101289. [PMID: 31640251 PMCID: PMC6830076 DOI: 10.3390/cells8101289] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 01/20/2023] Open
Abstract
Charcot–Marie tooth disease is a hereditary polyneuropathy caused by mutations in Mitofusin-2 (MFN2), a GTPase in the outer mitochondrial membrane involved in the regulation of mitochondrial fusion and bioenergetics. Autosomal-dominant inheritance of a R94Q mutation in MFN2 causes the axonal subtype 2A2A which is characterized by early onset and progressive atrophy of distal muscles caused by motoneuronal degeneration. Here, we studied mitochondrial shape, respiration, cytosolic, and mitochondrial ATP content as well as mitochondrial quality control in MFN2-deficient fibroblasts stably expressing wildtype or R94Q MFN2. Under normal culture conditions, R94Q cells had slightly more fragmented mitochondria but a similar mitochondrial oxygen consumption, membrane potential, and ATP production as wildtype cells. However, when inducing mild oxidative stress 24 h before analysis using 100 µM hydrogen peroxide, R94Q cells exhibited significantly increased respiration but decreased mitochondrial ATP production. This was accompanied by increased glucose uptake and an up-regulation of hexokinase 1 and pyruvate kinase M2, suggesting increased pyruvate shuttling into mitochondria. Interestingly, these changes coincided with decreased levels of PINK1/Parkin-mediated mitophagy in R94Q cells. We conclude that mitochondria harboring the disease-causing R94Q mutation in MFN2 are more susceptible to oxidative stress, which causes uncoupling of respiration and ATP production possibly by a less efficient mitochondrial quality control.
Collapse
Affiliation(s)
- Christina Wolf
- Institute of Molecular Medicine, University Medical Center, Johannes Gutenberg-Universität Mainz, 55131 Mainz, Germany.
| | - Rahel Zimmermann
- Institute of Molecular Medicine, University Medical Center, Johannes Gutenberg-Universität Mainz, 55131 Mainz, Germany.
| | - Osamah Thaher
- Institute of Molecular Medicine, University Medical Center, Johannes Gutenberg-Universität Mainz, 55131 Mainz, Germany.
| | - Diones Bueno
- Institute of Molecular Medicine, University Medical Center, Johannes Gutenberg-Universität Mainz, 55131 Mainz, Germany.
| | - Verena Wüllner
- Institute of Molecular Medicine, University Medical Center, Johannes Gutenberg-Universität Mainz, 55131 Mainz, Germany.
| | - Michael K E Schäfer
- Department of Anesthesiology, Research Center for Immunotherapy (FZI), Focus Program Translational Neurosciences (FTN), University Medical Center, Johannes Gutenberg-Universität Mainz, 55116 Mainz, Germany.
| | - Philipp Albrecht
- Department of Neurology, University Hospital Düsseldorf, 40210 Düsseldorf, Germany.
| | - Axel Methner
- Institute of Molecular Medicine, University Medical Center, Johannes Gutenberg-Universität Mainz, 55131 Mainz, Germany.
| |
Collapse
|
20
|
Chakraborty D, Felzen V, Hiebel C, Stürner E, Perumal N, Manicam C, Sehn E, Grus F, Wolfrum U, Behl C. Enhanced autophagic-lysosomal activity and increased BAG3-mediated selective macroautophagy as adaptive response of neuronal cells to chronic oxidative stress. Redox Biol 2019; 24:101181. [PMID: 30959460 PMCID: PMC6454062 DOI: 10.1016/j.redox.2019.101181] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 01/05/2023] Open
Abstract
Oxidative stress and a disturbed cellular protein homeostasis (proteostasis) belong to the most important hallmarks of aging and of neurodegenerative disorders. The proteasomal and autophagic-lysosomal degradation pathways are key measures to maintain proteostasis. Here, we report that hippocampal cells selected for full adaptation and resistance to oxidative stress induced by hydrogen peroxide (oxidative stress-resistant cells, OxSR cells) showed a massive increase in the expression of components of the cellular autophagic-lysosomal network and a significantly higher overall autophagic activity. A comparative expression analysis revealed that distinct key regulators of autophagy are upregulated in OxSR cells. The observed adaptive autophagic response was found to be independent of the upstream autophagy regulator mTOR but is accompanied by a significant upregulation of further downstream components of the canonical autophagy network such as Beclin1, WIPI1 and the transmembrane ATG9 proteins. Interestingly, the expression of the HSP70 co-chaperone BAG3, mediator of BAG3-mediated selective macroautophagy and highly relevant for the clearance of aggregated proteins in cells, was found to be increased in OxSR cells that were consequently able to effectively overcome proteotoxic stress. Overexpression of BAG3 in oxidative stress-sensitive HT22 wildtype cells partly established the vesicular phenotype and the enhanced autophagic flux seen in OxSR cells suggesting that BAG3 takes over an important part in the adaptation process. A full proteome analysis demonstrated additional changes in the expression of mitochondrial proteins, metabolic enzymes and different pathway regulators in OxSR cells as consequence of the adaptation to oxidative stress in addition to autophagy-related proteins. Taken together, this analysis revealed a wide variety of pathways and players that act as adaptive response to chronic redox stress in neuronal cells.
Collapse
Affiliation(s)
- Debapriya Chakraborty
- Institute of Pathobiochemistry, University Medical Center Mainz of the Johannes Gutenberg University, 55099, Mainz, Germany.
| | - Vanessa Felzen
- Institute of Pathobiochemistry, University Medical Center Mainz of the Johannes Gutenberg University, 55099, Mainz, Germany.
| | - Christof Hiebel
- Institute of Pathobiochemistry, University Medical Center Mainz of the Johannes Gutenberg University, 55099, Mainz, Germany.
| | - Elisabeth Stürner
- Institute of Pathobiochemistry, University Medical Center Mainz of the Johannes Gutenberg University, 55099, Mainz, Germany.
| | - Natarajan Perumal
- Experimental and Translational Ophthalmology, University Medical Center Mainz, 55131, Mainz, Germany.
| | - Caroline Manicam
- Experimental and Translational Ophthalmology, University Medical Center Mainz, 55131, Mainz, Germany.
| | - Elisabeth Sehn
- Institute for Molecular Physiology, Johannes Gutenberg University, 55128, Mainz, Germany.
| | - Franz Grus
- Experimental and Translational Ophthalmology, University Medical Center Mainz, 55131, Mainz, Germany.
| | - Uwe Wolfrum
- Institute for Molecular Physiology, Johannes Gutenberg University, 55128, Mainz, Germany.
| | - Christian Behl
- Institute of Pathobiochemistry, University Medical Center Mainz of the Johannes Gutenberg University, 55099, Mainz, Germany.
| |
Collapse
|
21
|
Hirata Y, Iwasaki T, Makimura Y, Okajima S, Oh-Hashi K, Takemori H. Inhibition of double-stranded RNA-dependent protein kinase prevents oxytosis and ferroptosis in mouse hippocampal HT22 cells. Toxicology 2019; 418:1-10. [PMID: 30817950 DOI: 10.1016/j.tox.2019.02.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/26/2019] [Accepted: 02/22/2019] [Indexed: 01/20/2023]
Abstract
Double-stranded RNA-dependent protein kinase (PKR) is a component of signal transduction pathways mediating various stress signals including oxidative stress and endoplasmic reticulum (ER) stress and is suggested to be implicated in several neurodegenerative diseases. Cell death in neurodegenerative conditions has been linked to oxidative stress; however, the involvement of PKR in endogenous oxidative stress such as oxytosis and ferroptosis which is quite distinct from classical apoptosis remains unknown. We investigated here the effect of a PKR inhibitor C16 (an imidazole-oxindole derivative) on oxytosis and ferroptosis in cultured HT22 mouse hippocampal cells. C16 prevented glutamate- and erastin-induced cell death, reactive oxygen species accumulation, Ca2+ influx, phosphorylation of inositol-requiring enzyme 1 (IRE1), one of the three branches of ER stress signaling and its downstream signaling components. On the other hand, C16 did not prevent oxidative stress-induced heme oxygenase-1 expression; instead, C16 activated the extracellular signal-regulated kinase pathway. The protective effect of C16 is diminished in PKR knockout HT22 cells. Real time measurements of the oxygen consumption rate and extracellular acidification rate over a long period of time leading to cell death showed that C16 partially prevented erastin-induced mitochondrial and glycolytic dysfunction. These results suggest that PKR is an important component of oxytosis and ferroptosis and the inhibition of PKR is neuroprotective against endogenous oxidative stress-induced cell death and provide an effective strategy for neuroprotection.
Collapse
Affiliation(s)
- Yoko Hirata
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido, Gifu 501-1193, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido, Gifu 501-1193, Japan.
| | - Takuya Iwasaki
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido, Gifu 501-1193, Japan
| | - Yukimi Makimura
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido, Gifu 501-1193, Japan
| | - Sayaka Okajima
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido, Gifu 501-1193, Japan
| | - Kentaro Oh-Hashi
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido, Gifu 501-1193, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido, Gifu 501-1193, Japan
| | - Hiroshi Takemori
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido, Gifu 501-1193, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido, Gifu 501-1193, Japan.
| |
Collapse
|
22
|
Zhang J, Feng J, Ma D, Wang F, Wang Y, Li C, Wang X, Yin X, Zhang M, Dagda RK, Zhang Y. Neuroprotective Mitochondrial Remodeling by AKAP121/PKA Protects HT22 Cell from Glutamate-Induced Oxidative Stress. Mol Neurobiol 2019; 56:5586-5607. [PMID: 30652267 DOI: 10.1007/s12035-018-1464-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/19/2018] [Indexed: 01/05/2023]
Abstract
Protein kinase A (PKA) is a ser/thr kinase that is critical for maintaining essential neuronal functions including mitochondrial homeostasis, bioenergetics, neuronal development, and neurotransmission. The endogenous pool of PKA is targeted to the mitochondrion by forming a complex with the mitochondrial scaffold A-kinase anchoring protein 121 (AKAP121). Enhanced PKA signaling via AKAP121 leads to PKA-mediated phosphorylation of the fission modulator Drp1, leading to enhanced mitochondrial networks and thereby blocking apoptosis against different toxic insults. In this study, we show for the first time that AKAP121/PKA confers neuroprotection in an in vitro model of oxidative stress induced by exposure to excess glutamate. Unexpectedly, treating mouse hippocampal progenitor neuronal HT22 cells with an acute dose or chronic exposure of glutamate robustly elevates PKA signaling, a beneficial compensatory response that is phenocopied in HT22 cells conditioned to thrive in the presence of excess glutamate but not in parental HT22 cells. Secondly, redirecting the endogenous pool of PKA by transiently transfecting AKAP121 or transfecting a constitutively active mutant of PKA targeted to the mitochondrion (OMM-PKA) or of an isoform of AKAP121 that lacks the KH and Tudor domains (S-AKAP84) are sufficient to significantly block cell death induced by glutamate toxicity but not in an oxygen deprivation/reperfusion model. Conversely, transient transfection of HT22 neuronal cells with a PKA-binding-deficient mutant of AKAP121 is unable to protect against oxidative stress induced by glutamate toxicity suggesting that the catalytic activity of PKA is required for AKAP121's protective effects. Mechanistically, AKAP121 promotes neuroprotection by enhancing PKA-mediated phosphorylation of Drp1 to increase mitochondrial fusion, elevates ATP levels, and elicits an increase in the levels of antioxidants GSH and superoxide dismutase 2 leading to a reduction in the level of mitochondrial superoxide. Overall, our data supports AKAP121/PKA as a new molecular target that confers neuroprotection against glutamate toxicity by phosphorylating Drp1, to stabilize mitochondrial networks and mitochondrial function and to elicit antioxidant responses.
Collapse
Affiliation(s)
- Jingdian Zhang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Feng Wang
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yumeng Wang
- Department of Physiology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Xinmin Street No. 126, Changchun, 130000, China
| | - Chunxiao Li
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Xiang Yin
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Ruben K Dagda
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Mailstop 318, Howard Medical Sciences Building 148A (Office), Reno, NV, 89557,, USA
| | - Ying Zhang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China.
| |
Collapse
|
23
|
Liu H, Liu J, Si L, Guo C, Liu W, Liu Y. GDF-15 promotes mitochondrial function and proliferation in neuronal HT22 cells. J Cell Biochem 2019; 120:10530-10547. [PMID: 30635935 DOI: 10.1002/jcb.28339] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/11/2018] [Indexed: 01/21/2023]
Abstract
The neuronal cell line HT22 is an excellent model for studying Parkinson's disease. Growth differentiation factor 15 (GDF15) plays a critical role in Parkinson's disease, but the molecular mechanism involved are not well understood. We constructed the GDF15 overexpression HT22 cells and detected the effects of overexpression of GDF15 on the viability, oxygen consumption, mitochondrial membrane potential of oligomycin-treated HT22 cells. In addition, we used a high-throughput RNA-sequencing to study the lncRNA and mRNA expression profiling and obtained key lncRNAs, mRNA, gene ontology (GO), and Kyoto encyclopedia of genes and genomes (KEGG) pathway. The expression of selected DElncRNAs was validated by quantitative real-time PCR (qRT-PCR). Our results showed that overexpression of GDF15 significantly reversed the cells viability, oxygen consumption, and mitochondrial membrane potential effect caused by oligomycin in HT22 cells. The 1093 DEmRNAs and 395 DElncRNAs in HT22 cells between GDF15-oligomycin non-intervention group and a normal control-oligomycin un-intervention group were obtained, and 394 DEmRNAs and 271 DElncRNAs in HT22 cells between GDF15-oligomycin intervention group and normal control-oligomycin intervention group were identified. Base on the GO and KEGG enrichment analysis of between GDF15-oligomycin intervention group and normal control-oligomycin intervention group, positive regulation of cell proliferation was most significantly enriched GO terms, and Cav1 was enriched in positive regulation of cell proliferation pathway. PI3K-Akt signaling pathway was one significantly enriched pathway in GDF15-oligomycin intervention group. The qRT-PCR results were consistent with RNA-sequencing, generally. GDF15 might promote mitochondrial function and proliferation of HT22 cells by regulating PI3K/Akt signaling pathway. Our study may be helpful in understanding the potential molecular mechanism of GDF15 in Parkinson's disease.
Collapse
Affiliation(s)
- Hong Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of Neurology, People's Hospital of Liaocheng Affiliated to Taishan Medical College, Liaocheng, Shandong, China
| | - Jiahui Liu
- Department of Neurology, Baotou Central Hospital, Baotou, Inner Mongolia, China
| | - Lei Si
- Department of Precision, People's Hospital of Liaocheng Affiliated to Taishan Medical College, Liaocheng, Shandong, China
| | - Cunju Guo
- Department of Neurology, People's Hospital of Liaocheng Affiliated to Taishan Medical College, Liaocheng, Shandong, China
| | - Wei Liu
- Department of Central Laboratory, People's Hospital of Liaocheng Affiliated to Taishan Medical College, Liaocheng, Shandong, China
| | - Yiming Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
24
|
Li H, Xin C, Zhang G, Han X, Qin W, Zhang CW, Yu C, Jing S, Li L, Huang W. A mitochondria-targeted two-photon fluorogenic probe for the dual-imaging of viscosity and H2O2 levels in Parkinson's disease models. J Mater Chem B 2019. [DOI: 10.1039/c9tb00576e] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Novel two-photon fluorogenic probe could simultaneously monitor changes in the mitochondrial viscosity and H2O2 levels using two different channels.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
| | - Chenqi Xin
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
| | - Gaobin Zhang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
| | - Xisi Han
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
| | - Wenjing Qin
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
| | - Cheng-wu Zhang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
| | - Changmin Yu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
| | - Su Jing
- School of Chemistry and Molecular Engineering
- Nanjing Tech University
- Nanjing
- P. R. China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Nanjing Tech University (NanjingTech)
- Nanjing
- P. R. China
- Shaanxi Institute of Flexible Electronics (SIFE)
| |
Collapse
|
25
|
Xu QH, Song BJ, Liu D, Chen YH, Zhou Y, Liu WB, Li H, Long TL, Zhang R, Liu W. The MKK7 inhibitor peptide GADD45β-I attenuates ER stress-induced mitochondrial dysfunction in HT22 cells: Involvement of JNK-Wnt pathway. Brain Res 2018; 1691:1-8. [PMID: 29684334 DOI: 10.1016/j.brainres.2018.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 03/29/2018] [Accepted: 04/12/2018] [Indexed: 12/17/2022]
Abstract
JNK, a member of the mitogen activated protein kinases (MAPKs) superfamily, plays a key role in cell death in many neurological disorders, but systemic inhibition of JNK has detrimental side effects. JNK can be regulated by two direct upstream kinases: MAPK kinase 4 (MKK4) and MAPK kinase 7 (MKK7). Here, we investigated the effect of GADD45β-I, a recently designed cell-permeable inhibitor peptide for MKK7, on endoplasmic reticulum (ER) stress-induced cytotoxicity in neuronal HT22 cells. We found that treatment with the ER stress inducer tunicamycin (TM) increased the phosphorylation of JNK and MKK7 in HT22 cells, which was nullified by GADD45β-I. GADD45β-I significantly attenuated TM-induced toxicity via inhibiting apoptotic cell death, as evidenced by decreased number of TUNEL-positive cells and reduced caspase-3 activity. GADD45β-I treatment also decreased expression of ER stress associated pro-apoptotic proteins and prevented morphological changes of the ER after TM exposure. In addition, inhibition of mitochondrial oxidative stress and preservation of intracellular ATP levels were observed in GADD45β-I-treated cells. The experiments using siRNA transfection and Topflash reporter assay revealed a possible involvement of Wnt/β-catenin pathway in GADD45β-I-induced protection in HT22 cells. In summary, our results demonstrated that GADD45β-I exerted protective effects against TM-induced cytotoxicity via regulating JNK-Wnt pathway. Targeting MKK7 could represent a new therapeutic strategy for the treatment of neurological diseases where ER stress associated neuronal injury are involved.
Collapse
Affiliation(s)
- Quan-Hua Xu
- Department of Neurosurgery, Bijie First People's Hospital, Bijie, Guizhou 551700, China
| | - Bing-Jun Song
- Basic Medical Sciences Research Center, Shaanxi Fourth People's Hospital, Xi'an, Shaanxi 710043, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yu-Hua Chen
- Basic Medical Sciences Research Center, Shaanxi Fourth People's Hospital, Xi'an, Shaanxi 710043, China
| | - Yuan Zhou
- Basic Medical Sciences Research Center, Shaanxi Fourth People's Hospital, Xi'an, Shaanxi 710043, China
| | - Wen-Bo Liu
- Basic Medical Sciences Research Center, Shaanxi Fourth People's Hospital, Xi'an, Shaanxi 710043, China
| | - Hua Li
- Basic Medical Sciences Research Center, Shaanxi Fourth People's Hospital, Xi'an, Shaanxi 710043, China
| | - Tian-Lin Long
- Department of Neurosurgery, Bijie First People's Hospital, Bijie, Guizhou 551700, China
| | - Rui Zhang
- Department of Neurosurgery, Bijie First People's Hospital, Bijie, Guizhou 551700, China
| | - Wei Liu
- Basic Medical Sciences Research Center, Shaanxi Fourth People's Hospital, Xi'an, Shaanxi 710043, China.
| |
Collapse
|
26
|
Lewerenz J, Ates G, Methner A, Conrad M, Maher P. Oxytosis/Ferroptosis-(Re-) Emerging Roles for Oxidative Stress-Dependent Non-apoptotic Cell Death in Diseases of the Central Nervous System. Front Neurosci 2018; 12:214. [PMID: 29731704 PMCID: PMC5920049 DOI: 10.3389/fnins.2018.00214] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/19/2018] [Indexed: 12/12/2022] Open
Abstract
Although nerve cell death is the hallmark of many neurological diseases, the processes underlying this death are still poorly defined. However, there is a general consensus that neuronal cell death predominantly proceeds by regulated processes. Almost 30 years ago, a cell death pathway eventually named oxytosis was described in neuronal cells that involved glutathione depletion, reactive oxygen species production, lipoxygenase activation, and calcium influx. More recently, a cell death pathway that involved many of the same steps was described in tumor cells and termed ferroptosis due to a dependence on iron. Since then there has been a great deal of discussion in the literature about whether these are two distinct pathways or cell type- and insult-dependent variations on the same pathway. In this review, we compare and contrast in detail the commonalities and distinctions between the two pathways concluding that the molecular pathways involved in the regulation of ferroptosis and oxytosis are highly similar if not identical. Thus, we suggest that oxytosis and ferroptosis should be regarded as two names for the same cell death pathway. In addition, we describe the potential physiological relevance of oxytosis/ferroptosis in multiple neurological diseases.
Collapse
Affiliation(s)
- Jan Lewerenz
- Department of Neurology, Ulm University, Ulm, Germany
| | - Gamze Ates
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Axel Methner
- Department of Neurology, University Medical Center and Focus Program Translational Neuroscience of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
27
|
Hsu WH, Lin YC, Chen BR, Wu SC, Lee BH. The neuronal protection of a zinc-binding protein isolated from oyster. Food Chem Toxicol 2018; 114:61-68. [PMID: 29432843 DOI: 10.1016/j.fct.2018.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/22/2018] [Accepted: 02/06/2018] [Indexed: 12/19/2022]
Abstract
Mitochondrial function is applied as oxidative stress and neuronal damage index. In this study, d-galactose was used to induce free radicals production and neuronal damage in HN-h cells, and the effect of novel 43 kDa protein isolated from oyster on anti-mitochondrial dysfunction and zinc-binding ability were evaluated. Crystal violet stain results indicated zinc-binding protein of oyster (ZPO) attenuated neuronal cell death induced by 100 mM of d-galactose on HN-h cells in a dose-dependent manner. ZPO alleviated mitochondrial inactivation, mitochondrial membrane potential decreasing, oxidative stress, and fusion/fission state in non-cytotoxic concentration of d-galactose (50 mM)-treated HN-h cells. ZPO treatment recovered metallathionein-3 (MT-3) decrease and inhibited β- and γ-secretase as well as amyloid beta (Aβ) accumulation in HN-h cells caused by d-galactose induction. These results suggest ZPO could avoid oxidative stress and is a functional protein for zinc concentration maintainability, which has potential for development of functional foods for neuronal protection.
Collapse
Affiliation(s)
- Wei-Hsuan Hsu
- Biochemical Process Technology Department, Center of Excellence for Drug Development, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan, ROC
| | - Yu-Chun Lin
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | - Bo-Rui Chen
- Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan, ROC; Doctoral Degree Program in Marine Biotechnology, National Taiwan Ocean University, Keelung, Taiwan, ROC; Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan, ROC
| | - She-Ching Wu
- Department of Food Science, National Chiayi University, Chiayi, Taiwan, ROC.
| | - Bao-Hong Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan, ROC; Department of Chinese Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan, ROC.
| |
Collapse
|
28
|
Larrick JW, Larrick JW, Mendelsohn AR. ATP Synthase, a Target for Dementia and Aging? Rejuvenation Res 2018; 21:61-66. [DOI: 10.1089/rej.2018.2056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- James W. Larrick
- Panorama Research Institute, Sunnyvale, California
- Regenerative Sciences Institute, Sunnyvale, California
| | | | - Andrew R. Mendelsohn
- Panorama Research Institute, Sunnyvale, California
- Regenerative Sciences Institute, Sunnyvale, California
| |
Collapse
|
29
|
Pfeiffer A, Schneider J, Bueno D, Dolga A, Voss TD, Lewerenz J, Wüllner V, Methner A. Bcl-x L knockout attenuates mitochondrial respiration and causes oxidative stress that is compensated by pentose phosphate pathway activity. Free Radic Biol Med 2017; 112:350-359. [PMID: 28807815 DOI: 10.1016/j.freeradbiomed.2017.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 08/01/2017] [Accepted: 08/08/2017] [Indexed: 12/12/2022]
Abstract
Bcl-xL is an anti-apoptotic protein that localizes to the outer mitochondrial membrane and influences mitochondrial bioenergetics by controlling Ca2+ influx into mitochondria. Here, we analyzed the effect of mitochondrial Bcl-xL on mitochondrial shape and function in knockout (KO), wild type and rescued mouse embryonic fibroblast cell lines. Mitochondria of KO cells were more fragmented, exhibited a reduced ATP concentration, and reduced oxidative phosphorylation (OXPHOS) suggesting an increased importance of ATP generation by other means. Under steady-state conditions, acidification of the growth medium as a readout for glycolysis was similar, but upon inhibition of ATP synthase with oligomycin, KO cells displayed an instant increase in glycolysis. In addition, forced energy production through OXPHOS by replacing glucose with galactose in the growth medium rendered KO cells more susceptible to mitochondrial toxins. KO cells had increased cellular reactive oxygen species and were more susceptible to oxidative stress, but had higher glutathione levels, which were however more rapidly consumed under conditions of oxidative stress. This coincided with an increased activity and protein abundance of the pentose phosphate pathway protein glucose-6-phosphate dehydrogenase, which generates NADPH necessary to regenerate reduced glutathione. KO cells were also less susceptible to pharmacological inhibition of the pentose phosphate pathway. We conclude that mitochondrial Bcl-xL is involved in maintaining mitochondrial respiratory capacity. Its deficiency causes oxidative stress, which is associated with an increased glycolytic capacity and balanced by an increased activity of the pentose phosphate pathway.
Collapse
Affiliation(s)
- Annika Pfeiffer
- Department of Neurology, University Medical Center and Focus Program Translational Neuroscience (FTN) of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Julia Schneider
- Department of Neurology, University Medical Center and Focus Program Translational Neuroscience (FTN) of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Diones Bueno
- Department of Neurology, University Medical Center and Focus Program Translational Neuroscience (FTN) of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Amalia Dolga
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Timo-Daniel Voss
- Universitäts, und Rehabilitationskliniken Ulm, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Jan Lewerenz
- Universitäts, und Rehabilitationskliniken Ulm, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Verena Wüllner
- Department of Neurology, University Medical Center and Focus Program Translational Neuroscience (FTN) of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Axel Methner
- Department of Neurology, University Medical Center and Focus Program Translational Neuroscience (FTN) of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
30
|
Zhou ZR, Huang P, Song GH, Zhang Z, An K, Lu HW, Ju XL, Ding W. Comparative proteomic analysis of rats subjected to water immersion and restraint stress as an insight into gastric ulcers. Mol Med Rep 2017; 16:5425-5433. [PMID: 28849061 PMCID: PMC5647087 DOI: 10.3892/mmr.2017.7241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 06/15/2017] [Indexed: 12/13/2022] Open
Abstract
In the present study, comparative proteomic analysis was performed in rats subjected to water immersion-restraint stress (WRS). A total of 26 proteins were differentially expressed and identified using matrix-assisted laser desorption/ionization time of flight mass spectrometry. Among the 26 differentially expressed protein spots identified, 13 proteins were significantly upregulated under WRS, including pyruvate kinase and calreticulin, which may be closely associated with energy metabolism. In addition, 12 proteins were downregulated under WRS, including hemoglobin subunit β-2 and keratin type II cytoskeletal 8, which may be important in protein metabolism and cell death. Gene Ontology analysis revealed the cellular distribution, molecular function and biological processes of the identified proteins. The mRNA levels of certain differentially expressed proteins were analyzed using fluorescence quantitative polymerase chain reaction analysis. The results of the present study aimed to offer insights into proteins, which are differentially expressed in gastric ulcers in stress, and provide theoretical evidence of a radical cure for gastric ulcers in humans.
Collapse
Affiliation(s)
- Zheng-Rong Zhou
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Pan Huang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Guang-Hao Song
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhuang Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ke An
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Han-Wen Lu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiao-Li Ju
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wei Ding
- Department of Animal Husbandry and Veterinary Medicine, Jiangsu Polytechnic College of Agriculture and Forestry, Jurong, Jiangsu 212499, P.R. China
| |
Collapse
|
31
|
Maher P, van Leyen K, Dey PN, Honrath B, Dolga A, Methner A. The role of Ca 2+ in cell death caused by oxidative glutamate toxicity and ferroptosis. Cell Calcium 2017; 70:47-55. [PMID: 28545724 DOI: 10.1016/j.ceca.2017.05.007] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022]
Abstract
Ca2+ ions play a fundamental role in cell death mediated by oxidative glutamate toxicity or oxytosis, a form of programmed cell death similar and possibly identical to other forms of cell death like ferroptosis. Ca2+ influx from the extracellular space occurs late in a cascade characterized by depletion of the intracellular antioxidant glutathione, increases in cytosolic reactive oxygen species and mitochondrial dysfunction. Here, we aim to compare oxidative glutamate toxicity with ferroptosis, address the signaling pathways that culminate in Ca2+ influx and cell death and discuss the proteins that mediate this. Recent evidence hints toward a role of the machinery responsible for store-operated Ca2+ entry (SOCE), which refills the endoplasmic reticulum (ER) after receptor-mediated ER Ca2+ release or other forms of store depletion. Pharmacological inhibition of SOCE or transcriptional downregulation of proteins involved in SOCE like the ER Ca2+ sensor STIM1, the plasma membrane Ca2+ channels Orai1 and TRPC1 and the linking protein Homer protects against oxidative glutamate toxicity and direct oxidative stress caused by hydrogen peroxide or 1-methyl-4-phenylpyridinium (MPP+) injury, a cellular model of Parkinson's disease. This suggests that SOCE inhibition might have some potential therapeutic effects in human disease associated with oxidative stress like neurodegenerative disorders.
Collapse
Affiliation(s)
- Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Partha Narayan Dey
- University Medical Center and Focus Program Translational Neuroscience (FTN) of the Johannes Gutenberg University Mainz, Department of Neurology, Mainz, Germany
| | - Birgit Honrath
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Amalia Dolga
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Axel Methner
- University Medical Center and Focus Program Translational Neuroscience (FTN) of the Johannes Gutenberg University Mainz, Department of Neurology, Mainz, Germany.
| |
Collapse
|
32
|
Zimmermann L, Moldzio R, Vazdar K, Krewenka C, Pohl EE. Nutrient deprivation in neuroblastoma cells alters 4-hydroxynonenal-induced stress response. Oncotarget 2017; 8:8173-8188. [PMID: 28030790 PMCID: PMC5352392 DOI: 10.18632/oncotarget.14132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/21/2016] [Indexed: 01/29/2023] Open
Abstract
4-hydroxy-2-nonenal (HNE), a toxic lipid peroxidation product, is associated with oxidative damage in cells and involved in various diseases including the initiation and progression of cancer. Cancer cells have a high, adaptable metabolism with a shift from oxidative phosphorylation to glycolysis and rely on high levels of glucose and glutamine as essential nutrients for cell growth. Here we investigated whether the toxic effects of HNE on the mitochondrial membrane potential (MMP) of cancer cells depends on their metabolic state by deprivation of glucose and/or glutamine. The addition of 16 μM HNE to N18TG2 neuroblastoma cells incubated in glucose medium led to a severe reduction of MMP, which was similar to the MMP of cells fed with both glucose and glutamine. In contrast, HNE addition to cells starved in glutamine medium increased their MMP slightly for a prolonged time period and this was accompanied by increased cellular survival. We found that ß-oxidation of HNE did not cause the increased MMP, since the aldehyde dehydrogenase was distinctly more active in cells with glucose medium. However, after blocking fatty acid ß-oxidation in cells starved in glutamine medium with etomoxir, which inhibits carnitine palmitoyltransferase 1, HNE addition induced a strong reduction of MMP similar to cells in glucose medium. Surprisingly, the effect of more toxic 4-oxo-2-nonenal was less pronounced. Our results suggest that in contrast to cells fed with glucose, glutamine-fed cancer cells are capable of ß-oxidizing fatty acids to maintain their MMP to combat the toxic effects of HNE.
Collapse
Affiliation(s)
- Lars Zimmermann
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Rudolf Moldzio
- Institute of Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Katarina Vazdar
- Division of Organic Chemistry and Biochemistry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Christopher Krewenka
- Institute of Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Elena E. Pohl
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
33
|
Gen-27, a newly synthesized flavonoid, inhibits glycolysis and induces cell apoptosis via suppression of hexokinase II in human breast cancer cells. Biochem Pharmacol 2016; 125:12-25. [PMID: 27818240 DOI: 10.1016/j.bcp.2016.11.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 11/01/2016] [Indexed: 12/16/2022]
Abstract
We have previously reported that Gen-27, a newly synthesized flavonoid, exhibits anticancer effects against human colorectal cancer cells. In this study, we investigated the anticancer effects in human breast cancer cell lines and its underlying mechanisms. We demonstrated that Gen-27 inhibited the growth and proliferation of human breast cancer cells in concentration and time-dependent manners. It was found that Gen-27 induced mitochondrial-mediated apoptosis, characterized by the dissipation of mitochondrial membrane potential (ΔΨm), cytochrome c (Cyt c) release from mitochondria to cytosol, activation of caspases and induction of poly (ADP-ribose) polymerase (PARP). In addition, Gen-27 inhibited the glycolysis in human breast cancer cells. After treatment with Gen-27, the expression of HKII was down-regulated, accompanied by weakened interaction of HKII and VDAC. Further research revealed that the induction of mitochondrial apoptosis was associated with the decrease of HKII expression by Gen-27. Finally, in vivo studies demonstrated that Gen-27 significantly suppressed the growth and promoted apoptosis of MDA-MB-231 breast cancer orthotopic tumors with low systemic toxicity. In conclusion, the results showed that Gen-27 had significant anticancer effects against human breast cancer and it may potentially be used as a novel anticancer agent for the treatment of breast cancer.
Collapse
|
34
|
Wang B, Liu H, Yue L, Li X, Zhao L, Yang X, Wang X, Yang Y, Qu Y. Neuroprotective effects of pterostilbene against oxidative stress injury: Involvement of nuclear factor erythroid 2-related factor 2 pathway. Brain Res 2016; 1643:70-9. [DOI: 10.1016/j.brainres.2016.04.048] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 10/21/2022]
|
35
|
FAM3A attenuates ER stress-induced mitochondrial dysfunction and apoptosis via CHOP-Wnt pathway. Neurochem Int 2016; 94:82-9. [DOI: 10.1016/j.neuint.2016.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 12/13/2022]
|
36
|
Tu YL, Chen QH, Wang SN, Uri A, Yang XH, Chu JQ, Chen JK, Luo BL, Chen XH, Wen SJ, Pi RB. Discovery of lipoic acid-4-phenyl-1H-pyrazole hybrids as novel bifunctional ROCK inhibitors with antioxidant activity. RSC Adv 2016. [DOI: 10.1039/c6ra12081d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A potently selective ROCK2 inhibitor with antioxidative properties.
Collapse
|
37
|
Sun J, Ren X, Simpkins JW. Sequential Upregulation of Superoxide Dismutase 2 and Heme Oxygenase 1 by tert-Butylhydroquinone Protects Mitochondria during Oxidative Stress. Mol Pharmacol 2015; 88:437-49. [PMID: 26082377 PMCID: PMC4551047 DOI: 10.1124/mol.115.098269] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/16/2015] [Indexed: 12/30/2022] Open
Abstract
Oxidative stress is linked to mitochondrial dysfunction in aging and neurodegenerative conditions. The transcription factor nuclear factor E2-related factor 2 (Nrf2)-antioxidant response element (ARE) regulates intracellular antioxidative capacity to combat oxidative stress. We examined the effect of tert-butylhydroquinone (tBHQ), an Nrf2-ARE signaling pathway inducer, on mitochondrial function during oxidative challenge in neurons. tBHQ prevented glutamate-induced cytotoxicity in an HT-22 neuronal cell line even with an 8-hour exposure delay. tBHQ blocked glutamate-induced intracellular reactive oxygen species (ROS) and mitochondrial superoxide accumulation. It also protected mitochondrial function under glutamate toxicity, including maintaining mitochondrial membrane potential, mitochondrial Ca(2+) hemostasis, and mitochondrial respiration. Glutamate-activated, mitochondria-mediated apoptosis was inhibited by tBHQ as well. In rat primary cortical neurons, tBHQ protected cells from both glutamate and buthionine sulfoximine toxicity. We found that tBHQ scavenged ROS and induced a rapid upregulation of superoxide dismutase 2 (SOD2) expression and a delayed upregulation of heme oxygenase 1 (HO-1) expression. In HT-22 cells with a knockdown of SOD2 expression, delayed treatment with tBHQ failed to prevent glutamate-induced cell death. Briefly, tBHQ rescues mitochondrial function by sequentially increasing SOD2 and HO-1 expression during glutamate-mediated oxidative stress. This study is the first to demonstrate the role of tBHQ in preserving mitochondrial function during oxidative challenge and provides a clinically relevant argument for using tBHQ against acute neuron-compromising conditions.
Collapse
Affiliation(s)
- Jiahong Sun
- Department of Physiology and Pharmacology, Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, West Virginia
| | - Xuefang Ren
- Department of Physiology and Pharmacology, Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, West Virginia
| | - James W Simpkins
- Department of Physiology and Pharmacology, Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
38
|
Zhou Y, Lu N, Qiao C, Ni T, Li Z, Yu B, Guo Q, Wei L. FV-429 induces apoptosis and inhibits glycolysis by inhibiting Akt-mediated phosphorylation of hexokinase II in MDA-MB-231 cells. Mol Carcinog 2015; 55:1317-28. [PMID: 26258875 DOI: 10.1002/mc.22374] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 06/28/2015] [Accepted: 07/06/2015] [Indexed: 11/06/2022]
Abstract
In this study, the anticancer effect of a newly synthesized flavonoid FV-429, against human breast cancer MDA-MB-231 cells, and the underlying mechanisms were investigated. FV-429 triggered the apoptosis and simultaneously inhibited the glycolysis of MDA-MB-231 cells. Both the HK II activity and its level in mitochondria were significantly down regulated by FV-429. Moreover, FV-429 weakened the interaction between HKII and VDAC, stimulated the detachment of HK II from the mitochondria, and resulted in the opening of the mitochondrial permeability transition pores. Thus FV-429 induced the mitochondrial-mediated apoptosis, showing increased Bax/Bcl-2 ratio, loss of mitochondrial membrane potential (MMP) and activation of caspase-3 and -9, cytochrome c (Cyt c) release, and apoptosis inducing factor (AIF) transposition. Further research revealed that the phosphorylation of mitochondrial HKII via Akt was responsible for the dissociation of HKII and the decreased HKII activity induced by FV-429. Taken together, FV-429 inhibited the phosphorylation of HKII, down-regulated its activity, and stimulated the release of HKII from the mitochondria, resulting the inhibited glycolysis and mitochondrial-mediated apoptosis. The studies provide a molecular basis for the development of flavonoid compounds as novel anticancer agents for breast cancer. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, P. R. China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, P. R. China
| | - Chen Qiao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, P. R. China
| | - Ting Ni
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, P. R. China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, P. R. China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, 639# Long Mian Avenue, Nan Jing, P.R. China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, P. R. China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, JiangSu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, P. R. China
| |
Collapse
|
39
|
Leyton L, Hott M, Acuña F, Caroca J, Nuñez M, Martin C, Zambrano A, Concha MI, Otth C. Nutraceutical activators of AMPK/Sirt1 axis inhibit viral production and protect neurons from neurodegenerative events triggered during HSV-1 infection. Virus Res 2015; 205:63-72. [DOI: 10.1016/j.virusres.2015.05.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/20/2015] [Accepted: 05/20/2015] [Indexed: 11/29/2022]
|
40
|
Gannouni N, Lenoir M, Ben Rhouma K, El May M, Tebourbi O, Puel JL, Mhamdi A. Cochlear neuropathy in the rat exposed for a long period to moderate-intensity noises. J Neurosci Res 2015; 93:848-58. [DOI: 10.1002/jnr.23567] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/06/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Noura Gannouni
- Laboratory of Toxicology; Ergonomics and Occupational Environment, Faculty of Medicine of Tunis, Tunis El-Manar University; Tunis Tunisia
- Laboratory of Integrated Physiology; Faculty of Sciences of Bizerte; Carthage University; Tunis Tunisia
| | - Marc Lenoir
- INSERM Unit 1051. Deafness; Tinnitus and Therapies Research Unit. Institute of Neurosciences of Montpellier; Montpellier France
| | - Khemais Ben Rhouma
- Laboratory of Integrated Physiology; Faculty of Sciences of Bizerte; Carthage University; Tunis Tunisia
| | - Michèle El May
- Research Unit 01/UR/08-07, Department of Histology, Embryology and Cell Biology; Faculty of Medicine of Tunis, Tunis El-Manar University; Tunis Tunisia
| | - Olfa Tebourbi
- Laboratory of Integrated Physiology; Faculty of Sciences of Bizerte; Carthage University; Tunis Tunisia
| | - Jean Luc Puel
- INSERM Unit 1051. Deafness; Tinnitus and Therapies Research Unit. Institute of Neurosciences of Montpellier; Montpellier France
| | - Abada Mhamdi
- Laboratory of Toxicology; Ergonomics and Occupational Environment, Faculty of Medicine of Tunis, Tunis El-Manar University; Tunis Tunisia
| |
Collapse
|
41
|
Davidson SM, Lopaschuk GD, Spedding M, Beart PM. Mitochondrial pharmacology: energy, injury and beyond. Br J Pharmacol 2014; 171:1795-7. [PMID: 24684388 DOI: 10.1111/bph.12679] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
While the mitochondrion has long fascinated biologists and the sheer diversity of druggable targets has made it attractive for potential drug development, there has been little success translatable to the clinic. Given the diversity of inborn errors of metabolism and mitochondrial diseases, mitochondrially mediated oxidative stress (myopathies, reperfusion injury, Parkinson's disease, ageing) and the consequences of disturbed energetics (circulatory shock, diabetes, cancer), the potential for meaningful gain with novel drugs targeting mitochondrial mechanisms is huge both in terms of patient quality of life and health care costs. In this themed issue of the British Journal of Pharmacology, we highlight the key directions of the contemporary advances in the field of mitochondrial biology, emerging drug targets and new molecules which are close to clinical application. Authors' contributions are diverse both in terms of species and organs in which the mitochondrially related studies are performed, and from the perspectives of mechanisms under study. Defined roles of mitochondria in disease are updated and previously unknown contributions to disease are described in terms of the interface between basic science and pathological relevance.
Collapse
Affiliation(s)
- S M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | | | | | | |
Collapse
|