1
|
Mo S, Yang C, Zheng X, Lv H, Mao S, Liu N, Yang Q, Liao B, Yang M, Lu Z, Tang L, Huang X, Jian C, Li X, Shang J. Neuroprotective Effects of AER-271 in a tMCAO Mouse Model: Modulation of Autophagy, Apoptosis, and Inflammation. Inflammation 2024:10.1007/s10753-024-02082-7. [PMID: 39117789 DOI: 10.1007/s10753-024-02082-7] [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: 02/12/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024]
Abstract
Following ischemic stroke, aquaporin 4 (AQP4) expression modifications have been associated with increased inflammation. However, the underlying mechanisms are not fully understood. This study aims to elucidate the mechanistic basis of post-cerebral ischemia-reperfusion (I/R) inflammation by employing the AQP4-specific inhibitor, AER-271. The middle cerebral artery occlusion (MCAO) model was used to induce ischemic stroke in mice. C57BL/6 mice were randomly allocated into four groups: sham operation, I/R, AER-271, and 2-(nicotinamide)-1,3,4-thiadiazole (TGN-020) treatment, with observations recorded at 1 day, 3 days, and 7 days post-tMCAO. Each group consisted of 15 mice. Procedures included histological examination through HE staining, neurological scoring, Western blot analysis, and immunofluorescence staining. AER-271 treatment yielded significant improvements in post-stroke weight recovery and neurological scores, accompanied by a reduction in cerebral infarction volume. Moreover, AER-271 exhibited a noticeable influence on autophagic and apoptotic pathways, affecting the activation of both pro-inflammatory and anti-inflammatory cytokines. Alterations in the levels of inflammatory biomarkers MCP-1, NLRP3, and caspase 1 were also detected. Finally, a comparative assessment of the effects of AER-271 and TGN-020 in mitigating apoptosis and microglial polarization in ischemic mice revealed neuroprotective effects with no significant difference in efficacy. This study provides essential insights into the neuroprotective mechanisms of AER-271 in cerebral ischemia-reperfusion injury, offering potential clinical applications in the treatment of ischemic cerebrovascular disorders.
Collapse
Affiliation(s)
- Shenglong Mo
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Chengmin Yang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China
| | - Xingwu Zheng
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Hui Lv
- Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Sanyin Mao
- Department of Neurology, The First People's Hospital of Jiande, Hangzhou, China
| | - Ning Liu
- School of Basic Medical Sciences, Beihua University, Jilin, China
| | - Qin Yang
- Department of Neurology, BAISE PEOPLE'S HOSPITAL, Baise, Guangxi, China
| | - Bao Liao
- Department of Neurology, BAISE PEOPLE'S HOSPITAL, Baise, Guangxi, China
| | - Meiling Yang
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Zhicheng Lu
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Lina Tang
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Xiaorui Huang
- Department of Psychiatry and Psychology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chongdong Jian
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| | - Xuebin Li
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| | - Jingwei Shang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| |
Collapse
|
2
|
Moëlo C, Quillévéré A, Le Roy L, Timsit S. (S)-roscovitine, a CDK inhibitor, decreases cerebral edema and modulates AQP4 and α1-syntrophin interaction on a pre-clinical model of acute ischemic stroke. Glia 2024; 72:322-337. [PMID: 37828900 DOI: 10.1002/glia.24477] [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: 04/26/2023] [Revised: 08/22/2023] [Accepted: 09/07/2023] [Indexed: 10/14/2023]
Abstract
Cerebral edema is one of the deadliest complications of ischemic stroke for which there is currently no pharmaceutical treatment. Aquaporin-4 (AQP4), a water-channel polarized at the astrocyte endfoot, is known to be highly implicated in cerebral edema. We previously showed in randomized studies that (S)-roscovitine, a cyclin-dependent kinase inhibitor, reduced cerebral edema 48 h after induction of focal transient ischemia, but its mechanisms of action were unclear. In our recent blind randomized study, we confirmed that (S)-roscovitine was able to reduce cerebral edema by 65% at 24 h post-stroke (t test, p = .006). Immunofluorescence analysis of AQP4 distribution in astrocytes revealed that (S)-roscovitine decreased the non-perivascular pool of AQP4 by 53% and drastically increased AQP4 clusters in astrocyte perivascular end-feet (671%, t test p = .005) compared to vehicle. Non-perivascular and clustered AQP4 compartments were negatively correlated (R = -0.78; p < .0001), suggesting a communicating vessels effect between the two compartments. α1-syntrophin, AQP4 anchoring protein, was colocalized with AQP4 in astrocyte endfeet, and this colocalization was maintained in ischemic area as observed on confocal microscopy. Moreover, (S)-roscovitine increased AQP4/α1-syntrophin interaction (40%, MW p = .0083) as quantified by proximity ligation assay. The quantified interaction was negatively correlated with brain edema in both treated and placebo groups (R = -.57; p = .0074). We showed for the first time, that a kinase inhibitor modulated AQP4/α1-syntrophin interaction, and was implicated in the reduction of cerebral edema. These findings suggest that (S)-roscovitine may hold promise as a potential treatment for cerebral edema in ischemic stroke and as modulator of AQP4 function in other neurological diseases.
Collapse
Affiliation(s)
- Cloé Moëlo
- EFS, Université de Bretagne Occidentale, Inserm UMR 1078, GGB, Brest, France
| | - Alicia Quillévéré
- EFS, Université de Bretagne Occidentale, Inserm UMR 1078, GGB, Brest, France
| | - Lucas Le Roy
- EFS, Université de Bretagne Occidentale, Inserm UMR 1078, GGB, Brest, France
| | - Serge Timsit
- EFS, Université de Bretagne Occidentale, Inserm UMR 1078, GGB, Brest, France
- Neurology and Stroke Unit Department, CHRU de Brest, Inserm1078, Université de Bretagne Occidentale, Brest, France
| |
Collapse
|
3
|
Wang R, Wang M, Fan YC, Wang WJ, Zhang DH, Andy Li P, Zhang JZ, Jing L. Hyperglycemia exacerbates cerebral ischemia/reperfusion injury by up-regulating autophagy through p53-Sesn2-AMPK pathway. Neurosci Lett 2024; 821:137629. [PMID: 38191089 DOI: 10.1016/j.neulet.2024.137629] [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: 09/29/2023] [Revised: 12/14/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024]
Abstract
Hyperglycemia exacerbates ischemic brain injury by up-regulating autophagy. However, the underlying mechanisms are unknown. This study aims to determine whether hyperglycemia activates autophagy through the p53-Sesn2-AMPK signaling pathway. Rats were subjected to 30-min middle cerebral artery occlusion (MCAO) with reperfusion for 1- and 3-day under normo- and hyperglycemic conditions; and HT22 cells were exposed to oxygen deprivation (OG) or oxygen-glucose deprivation and re-oxygenation (OGD/R) with high glucose. Autophagy inhibitors, 3-MA and ARI, were used both in vivo and in vitro. The results showed that, compared with the normoglycemia group (NG), hyperglycemia (HG) increased infarct volume and apoptosis in penumbra area, worsened neurological deficit, and augmented autophagy. after MCAO followed by 1-day reperfusion. Further, HG promoted the conversion of LC-3I to LC-3II, decreased p62, increased protein levels of aldose reductase, p53, P-p53ser15, Sesn2, AMPK and numbers of autophagosomes and autolysosomes, detected by transmission electron microscopy and mRFP-GFP-LC3 molecular probe, in the cerebral cortex after ischemia and reperfusion injury in animals or in cultured HT22 cells exposed to hypoxia with high glucose content. Finally, experiments with autophagy inhibitors 3-MA and aldose reductase inhibitor (ARI) revealed that while both inhibitors reduced the number of TUNEL positive neurons and reversed the effects of hyperglycemic ischemia on LC3 and p62, only ARI decreased the levels of p53, P-p53ser15. These results suggested that hyperglycemia might induce excessive autophagy to aggravate the brain injury resulted from I/R and that hyperglycemia might activate the p53-Sesn2-AMPK signaling pathway, in addition to the classical PI3K/AKT/mTOR autophagy pathway.
Collapse
Affiliation(s)
- Rui Wang
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Meng Wang
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Yu-Cheng Fan
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Wen-Jun Wang
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Deng-Hai Zhang
- The Shanghai Health Commission Key Lab of Al-Based Management of Inflammation and Chronic Diseases, the Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Technology Enterprise, College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA
| | - Jian-Zhong Zhang
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| | - Li Jing
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
4
|
Llorián-Salvador M, Cabeza-Fernández S, Gomez-Sanchez JA, de la Fuente AG. Glial cell alterations in diabetes-induced neurodegeneration. Cell Mol Life Sci 2024; 81:47. [PMID: 38236305 PMCID: PMC10796438 DOI: 10.1007/s00018-023-05024-y] [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: 08/20/2023] [Revised: 10/09/2023] [Accepted: 10/29/2023] [Indexed: 01/19/2024]
Abstract
Type 2 diabetes mellitus is a global epidemic that due to its increasing prevalence worldwide will likely become the most common debilitating health condition. Even if diabetes is primarily a metabolic disorder, it is now well established that key aspects of the pathogenesis of diabetes are associated with nervous system alterations, including deleterious chronic inflammation of neural tissues, referred here as neuroinflammation, along with different detrimental glial cell responses to stress conditions and neurodegenerative features. Moreover, diabetes resembles accelerated aging, further increasing the risk of developing age-linked neurodegenerative disorders. As such, the most common and disabling diabetic comorbidities, namely diabetic retinopathy, peripheral neuropathy, and cognitive decline, are intimately associated with neurodegeneration. As described in aging and other neurological disorders, glial cell alterations such as microglial, astrocyte, and Müller cell increased reactivity and dysfunctionality, myelin loss and Schwann cell alterations have been broadly described in diabetes in both human and animal models, where they are key contributors to chronic noxious inflammation of neural tissues within the PNS and CNS. In this review, we aim to describe in-depth the common and unique aspects underlying glial cell changes observed across the three main diabetic complications, with the goal of uncovering shared glial cells alterations and common pathological mechanisms that will enable the discovery of potential targets to limit neuroinflammation and prevent neurodegeneration in all three diabetic complications. Diabetes and its complications are already a public health concern due to its rapidly increasing incidence, and thus its health and economic impact. Hence, understanding the key role that glial cells play in the pathogenesis underlying peripheral neuropathy, retinopathy, and cognitive decline in diabetes will provide us with novel therapeutic approaches to tackle diabetic-associated neurodegeneration.
Collapse
Affiliation(s)
- María Llorián-Salvador
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK.
| | - Sonia Cabeza-Fernández
- Institute for Health and Biomedical Research of Alicante (ISABIAL), Alicante, Spain
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Spain
| | - Jose A Gomez-Sanchez
- Institute for Health and Biomedical Research of Alicante (ISABIAL), Alicante, Spain
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Spain
| | - Alerie G de la Fuente
- Institute for Health and Biomedical Research of Alicante (ISABIAL), Alicante, Spain.
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Spain.
| |
Collapse
|