1
|
Song M, Zeng F, Huang L, Xiao H, Yi F, Sun X, Zheng L, Du H, Gu W. Energy restriction inhibits β-catenin ubiquitination to improve ischemic stroke injury via USP18/SKP2 axis. Metab Brain Dis 2024; 40:68. [PMID: 39692824 DOI: 10.1007/s11011-024-01494-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Abstract
Ischemic stroke (IS) remains a global health issue because of its great disability and mortality. Energy restriction (ER) has been justified to perform an inhibitory role in cerebral injury caused by IS. This research was purposed to inquire the potential molecular mechanism of ER in IS. To verify the function of ER in the animal and cell models of IS, rats were subjected to intermittent fasting (IF) and middle cerebral artery occlusion/reperfusion (MCAO/R) surgery and HAPI cells were treated with oxygen-glucose deprivation and reoxygenation (OGD/R) and 2-deoxyglucose (2-DG). It was disclosed that IF mitigated brain damage and inflammation in MCAO/R rats. Likewise, ER inhibited OGD/R-evoked microglial activation and inflammatory response. Of note, ubiquitin specific protease 18 (USP18) was uncovered to be the most significantly upregulated in MCAO/R rats receiving IF compared to free-feeding MCAO/R rats. Real-time quantitative polymerase chain reaction (RT-qPCR) and western blot verified that ER led to the promotion of USP18 expression. Moreover, downregulation of USP18 neutralized the meliorative effects of ER on OGD/R-treated HAPI cells. Functionally, USP18 restrained β-catenin ubiquitination to enhance its expression. In addition, our results manifested that S-phase kinase associated protein 2 (SKP2) contributed to degradation of β-catenin and USP18 abolished the role of SKP2 in β-catenin ubiquitination. Knockout of USP18 eliminated the protective effects of IF on MCAO/R rats, while SKP2 exacerbated brain damage and inflammation by decreasing β-catenin expression after IF. In summary, we validated that ER-induced USP18 exerts a suppressive function in IS damage through SKP2-mediated β-catenin ubiquitination.
Collapse
Affiliation(s)
- Mingyu Song
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China
| | - Feiyue Zeng
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Huang
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Hui Xiao
- Department of Neurology, Changsha Central Hospital, Changsha, Hunan, China
| | - Fang Yi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Geriatric neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinyu Sun
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China
| | - Lan Zheng
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China
| | - Hao Du
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China
| | - Wenping Gu
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China.
| |
Collapse
|
2
|
Zhang JF, Zhang L, Shi LL, Zhao ZH, Xu H, Liang F, Li HB, Zhao Y, Xu X, Yang K, Tian YF. Parthenolide attenuates cerebral ischemia/reperfusion injury via Akt/GSK-3β pathway in PC12 cells. Biomed Pharmacother 2017; 89:1159-1165. [PMID: 28314243 DOI: 10.1016/j.biopha.2017.03.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/28/2017] [Accepted: 03/03/2017] [Indexed: 11/19/2022] Open
Abstract
Parthenolide (PN), a sesquiterpene lactone isolated from the herbal medicine feverfew (Tanacetum parthenium), was reported to possess neuroprotective activity. However, the neuroprotective effect of PN against cerebral ischemia/reperfusion (I/R) injury remains unclear. Therefore, the aim of the present study was to explore the neuroprotective effects of PN against oxygen-glucose deprivation (OGD)-induced apoptosis in PC12 cells and the underlying mechanisms. Our results demonstrated that PN ameliorated OGD/R-evoked neuronal injury and oxidative stress in PC12 cells. In addition, PN notably decreased HIF-1α expression, as well as inhibited apoptosis in PC12 cells after OGD/R. Furthermore, PN pretreatment significantly enhanced the phosphorylation of Akt and GSK-3β in PC12 cells exposed to OGD/R. In conclusion, the present study demonstrated that PN exhibits a neuroprotective effect against OGD/R through activation of the Akt/GSK-3β signaling pathway. Our findings suggest that PN has the potential to serve as a novel therapeutic agent for cerebral I/R injury.
Collapse
Affiliation(s)
- Jun-Feng Zhang
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Li Zhang
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Li-Li Shi
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Zhao-Hua Zhao
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Hao Xu
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Fei Liang
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Hong-Bo Li
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Yan Zhao
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Xi Xu
- Department of Human Anatomy, Xi'an Medical University, Xi'an 710021, People's Republic of China.
| | - Ke Yang
- Department of Anesthesiology, The No.1 Hospital of Xi'an, Xi'an 710002, People's Republic of China.
| | - Ying-Fang Tian
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, People's Republic of China
| |
Collapse
|
3
|
Abstract
Although the protective mechanisms of delayed ischemic preconditioning have received extensive studies, few have addressed the mechanisms associated with rapid ischemic postconditioning. We investigated whether ischemic tolerance induced by rapid preconditioning is regulated by the Akt survival signaling pathway. Stroke was generated by permanent occlusion of the left distal middle cerebral artery (MCA) plus 30 min or 1 h occlusion of the bilateral common carotid artery (CCA) in male rats. Rapid preconditioning performed 1h before stroke onset reduced infarct size by 69% in rats with 30 min CCA occlusion, but by only 19% with 1 h occlusion. After control ischemia with 30 min CCA occlusion, Western Blot showed that P-Akt was transiently increased while Akt kinase assay showed that Akt activity was decreased. Although preconditioning did not change P-Akt levels at 1h and 5h compared with control ischemia, it attenuated reduction in Akt activity at 5h in the penumbra. However, preconditioning did not change the levels of P-PDK1, P-PTEN, and P-GSK3β in the Akt pathway, all of which were decreased after stroke. At last, the PI3K kinase inhibitor, LY294002, completely reversed the protection from ischemic preconditioning. In conclusion, Akt contributes to the protection of rapid preconditionin against stroke.
Collapse
|
4
|
Reduced beta-catenin expression in the hippocampal CA1 region following transient cerebral ischemia in the gerbil. Neurochem Res 2013; 38:1045-54. [PMID: 23504294 DOI: 10.1007/s11064-013-1015-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 02/08/2013] [Accepted: 03/05/2013] [Indexed: 12/16/2022]
Abstract
Beta-catenin, a transcription factor, plays a critical role in cell survival and degradation after stroke. In this study, we examined changes of expression in beta-catenin in the hippocampal CA1 region of the gerbil following 5 min of transient cerebral ischemia. We observed neuronal damage using cresyl violet staining, neuronal nuclei immunohistochemistry and Fluro-Jade B immunofluorescence. Four days after ischemia-reperfusion (I-R), most of pyramidal cells in the CA1 region were damaged. In addition, early damage in dendrites was detected 1 day after I-R by immunohistochemical staining for microtubule-associated protein 2 (MAP-2), and MAP-2 immunoreactivity was hardly detected in the CA1 region 4 days after I-R. We found that beta-catenin (a synapse-enriched cell adhesion molecule) was well expressed in dendrites before I-R. Its immunoreactivity was well colocalized with MAP-2. Chronological change of beta-catenin immunoreactivity was novelty in the present study. Twelve hours after I-R, its immunoreactivity was decreased in the stratum radiatum of the CA1 region, however, its immunoreactivity was increased 1 and 2 days after I-R, and decreased sharply 4 days after I-R. However, we did not find any change in beta-catenin immunoreactivity in the CA2 and CA3 region. In brief, we suggest that early change of beta-catenin expression in the stratum pyramidale of ischemic hippocampal CA1 region is associated with early dendrite damage following transient cerebral ischemia.
Collapse
|
5
|
Bhattacharya S, Chaum E, Johnson DA, Johnson LR. Age-related susceptibility to apoptosis in human retinal pigment epithelial cells is triggered by disruption of p53-Mdm2 association. Invest Ophthalmol Vis Sci 2012; 53:8350-66. [PMID: 23139272 DOI: 10.1167/iovs.12-10495] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Relatively little is known about the contribution of p53/Mdm2 pathway in apoptosis of retinal pigment epithelial (RPE) cells or its possible link to dysfunction of aging RPE or to related blinding disorders such as age-related macular degeneration (AMD). METHODS Age-associated changes in p53 activation were evaluated in primary RPE cultures from human donor eyes of various ages. Apoptosis was evaluated by activation of caspases and DNA fragmentation. Gene-specific small interfering RNA was used to knock down expression of p53. RESULTS We observed that the basal rate of p53-dependent apoptosis increased in an age-dependent manner in human RPE. The age-dependent increase in apoptosis was linked to alterations in several aspects of the p53 pathway. p53 phosphorylation Ser15 was increased through the stimulation of ATM-Ser1981. p53 acetylation Lys379 was increased through the inhibition of SIRT1/2. These two posttranslational modifications of p53 blocked the sequestration of p53 by Mdm2, thus resulting in an increase in free p53 and of p53 stimulation of apoptosis through increased expression of PUMA (p53 upregulated modulator of apoptosis) and activation of caspase-3. Aged RPE also had reduced expression of antiapoptotic Bcl-2, which contributed to the increase in apoptosis. Of particular interest in these studies was that pharmacologic treatments to block p53 phosphorylation, acetylation, or expression were able to protect RPE cells from apoptosis. CONCLUSIONS Our studies suggest that aging in the RPE leads to alterations of specific checkpoints in the apoptotic pathway, which may represent important molecular targets for the treatment of RPE-related aging disorders such as AMD.
Collapse
Affiliation(s)
- Sujoy Bhattacharya
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA.
| | | | | | | |
Collapse
|
6
|
Xing Y, Zhang X, Zhao K, Cui L, Wang L, Dong L, Li Y, Liu Z, Wang C, Zhang X, Zhu C, Qiao H, Ji Y, Cao X. Beneficial effects of sulindac in focal cerebral ischemia: a positive role in Wnt/β-catenin pathway. Brain Res 2012; 1482:71-80. [PMID: 22981403 DOI: 10.1016/j.brainres.2012.08.057] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 08/09/2012] [Accepted: 08/31/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND Accumulated evidences have established that inflammatory damage plays an important role in cerebral ischemic pathogenesis and may represent a target for treatment. Sulindac is well known as a nonsteroidal anti-inflammatory drug. However, little is known regarding the effect of sulindac in acute cerebral ischemia. Here, we designed this study to investigate the potential protective effects of sulindac in focal cerebral ischemia and the mechanisms underlying in vivo. METHODS Focal cerebral ischemia was induced in male Sprague-Dawley rats by permanent middle cerebral artery occlusion (pMCAO). Sulindac was administrated at dose of 4, 10, or 20mg/kg at 30 min before the operation. Neurological deficit scores, brain water content and infarct volumes were measured at 24h after pMCAO. Immunohistochemistry, western blot and reverse transcription-polymerase chain reaction were used for examining the mediators involved in Wnt/β-catenin signaling pathway, including the positive regulators dishevelled (Dvl) and β-catenin, the negative regulators adenomatous polyposis coli (APC), and P-β-catenin, as well as the downstream targets Bcl-2, Bax and claudin-5. RESULTS Compared with Vehicle group, 20mg/kg sulindac reduced neurological deficits, brain water content and infarct volumes. The same dose of sulindac upregulated the expression of Dvl, β-catenin, Bcl2 and claudin-5, and downregulated APC, P-β-catenin and Bax compared with Vehicle group. CONCLUSIONS These results showed that sulindac had a significant beneficial effect in cerebral ischemia; this effect may be correlated with the activation of the Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yinxue Xing
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Yang Y, Xia X, Zhang Y, Wang Q, Li L, Luo G, Xia Y. delta-Opioid receptor activation attenuates oxidative injury in the ischemic rat brain. BMC Biol 2009; 7:55. [PMID: 19709398 PMCID: PMC2754429 DOI: 10.1186/1741-7007-7-55] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 08/26/2009] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND We have recently shown that delta-opioid receptors (DORs) play an important role in neuroprotection from hypoxic injury via the regulation of extracellular signaling-regulated kinase (ERK) and cytochrome c release. Since ERK and cytochrome c are differentially involved in caspase signaling of oxidative injury that significantly contributes to neuronal damage in ischemia/reperfusion, we considered if DOR activation protects the ischemic brain by attenuating oxidative injury. RESULTS We observed that, in a model of cerebral ischemia with middle cerebral artery occlusion, DOR activation increased the activity of major antioxidant enzymes, glutathione peroxidase and superoxide dismutase, and decreased malondialdehyde and nitric oxide levels in the cortex exposed to cerebral ischemia/reperfusion. In addition, DOR activation reduced caspase 3 expression, though it did not significantly affect the increase in interleukin (IL)1beta and tumor necrosis factor (TNF)alpha expression at the same timepoint. PD98059, an inhibitor of mitogen-activated protein kinase (MAPK) extracellular signaling-regulated kinase kinase, accelerated animal death during ischemia/reperfusion. CONCLUSION DOR activation attenuates oxidative injury in the brain exposed to ischemia/reperfusion by enhancing antioxidant ability and inhibiting caspase activity, which provides novel insights into the mechanism of DOR neuroprotection.
Collapse
Affiliation(s)
- Yilin Yang
- Third Clinical College of Schoow University, Changzhou, Jiangsu, PR China
| | - Xiwei Xia
- Third Clinical College of Schoow University, Changzhou, Jiangsu, PR China
| | - Yi Zhang
- Third Clinical College of Schoow University, Changzhou, Jiangsu, PR China
| | - Qiang Wang
- Third Clinical College of Schoow University, Changzhou, Jiangsu, PR China
| | - Lu Li
- Third Clinical College of Schoow University, Changzhou, Jiangsu, PR China
| | - Guanghua Luo
- Third Clinical College of Schoow University, Changzhou, Jiangsu, PR China
| | - Ying Xia
- Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
8
|
Ren C, Gao X, Niu G, Yan Z, Chen X, Zhao H. Delayed postconditioning protects against focal ischemic brain injury in rats. PLoS One 2008; 3:e3851. [PMID: 19066627 PMCID: PMC2588536 DOI: 10.1371/journal.pone.0003851] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Accepted: 11/05/2008] [Indexed: 12/03/2022] Open
Abstract
Background We and others have reported that rapid ischemic postconditioning, interrupting early reperfusion after stroke, reduces infarction in rats. However, its extremely short therapeutic time windows, from a few seconds to minutes after reperfusion, may hinder its clinical translation. Thus, in this study we explored if delayed postconditioning, which is conducted a few hours after reperfusion, offers protection against stroke. Methods and Results Focal ischemia was generated by 30 min occlusion of bilateral common carotid artery (CCA) combined with permanent occlusion of middle cerebral artery (MCA); delayed postconditioning was performed by repetitive, brief occlusion and release of the bilateral CCAs, or of the ipsilateral CCA alone. As a result, delayed postconditioning performed at 3h and 6h after stroke robustly reduced infarct size, with the strongest protection achieved by delayed postconditioning with 6 cycles of 15 min occlusion/15 min release of the ipsilateral CCA executed from 6h. We found that this delayed postconditioning provided long-term protection for up to two months by reducing infarction and improving outcomes of the behavioral tests; it also attenuated reduction in 2-[18F]-fluoro-2-deoxy-D-glucose (FDG)-uptake therefore improving metabolism, and reduced edema and blood brain barrier leakage. Reperfusion in ischemic stroke patients is usually achieved by tissue plasminogen activator (tPA) application, however, t-PA's side effect may worsen ischemic injury. Thus, we tested whether delayed postconditioning counteracts the exacerbating effect of t-PA. The results showed that delayed postconditioning mitigated the worsening effect of t-PA on infarction. Conclusion Delayed postconditioning reduced ischemic injury after focal ischemia, which opens a new research avenue for stroke therapy and its underlying protective mechanisms.
Collapse
Affiliation(s)
- Chuancheng Ren
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- Department of Neurology, Shanghai No. 5 Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xuwen Gao
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
| | - Gang Niu
- Department of Radiology, Stanford University, Stanford, California, United States of America
| | - Zhimin Yan
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
| | - Xiaoyuan Chen
- Department of Radiology, Stanford University, Stanford, California, United States of America
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|