1
|
Wang H, Liu S, Sun Y, Chen C, Hu Z, Li Q, Long J, Yan Q, Liang J, Lin Y, Yang S, Lin M, Liu X, Wang H, Yu J, Yi F, Tan Y, Yang Y, Chen N, Ai Q. Target modulation of glycolytic pathways as a new strategy for the treatment of neuroinflammatory diseases. Ageing Res Rev 2024; 101:102472. [PMID: 39233146 DOI: 10.1016/j.arr.2024.102472] [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: 07/26/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/06/2024]
Abstract
Neuroinflammation is an innate and adaptive immune response initiated by the release of inflammatory mediators from various immune cells in response to harmful stimuli. While initially beneficial and protective, prolonged or excessive neuroinflammation has been identified in clinical and experimental studies as a key pathological driver of numerous neurological diseases and an accelerant of the aging process. Glycolysis, the metabolic process that converts glucose to pyruvate or lactate to produce adenosine 5'-triphosphate (ATP), is often dysregulated in many neuroinflammatory disorders and in the affected nerve cells. Enhancing glucose availability and uptake, as well as increasing glycolytic flux through pharmacological or genetic manipulation of glycolytic enzymes, has shown potential protective effects in several animal models of neuroinflammatory diseases. Modulating the glycolytic pathway to improve glucose metabolism and ATP production may help alleviate energy deficiencies associated with these conditions. In this review, we examine six neuroinflammatory diseases-stroke, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and depression-and provide evidence supporting the role of glycolysis in their treatment. We also explore the potential link between inflammation-induced aging and glycolysis. Additionally, we briefly discuss the critical role of glycolysis in three types of neuronal cells-neurons, microglia, and astrocytes-within physiological processes. This review highlights the significance of glycolysis in the pathology of neuroinflammatory diseases and its relevance to the aging process.
Collapse
Affiliation(s)
- Hanlong Wang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal&Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Ziyi Hu
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qinqin Li
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jinping Liang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xuan Liu
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Huiqin Wang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jingbo Yu
- Technology Innovation Center/National Key Laboratory Breeding Base of Chinese Medicine Powders and Innovative Drugs, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Fan Yi
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing 100048, China
| | - Yong Tan
- Nephrology Department, Xiangtan Central Hospital, Xiangtan 411100, China
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| |
Collapse
|
2
|
Yajun Z, Diqing O, Xingwei L, Liuyang T, Xiaofeng Z, Xiaoguo L, Zongduo G. High levels of blood lipid and glucose predict adverse prognosis in patients with aneurysmal subarachnoid hemorrhage. Heliyon 2024; 10:e38601. [PMID: 39397996 PMCID: PMC11470529 DOI: 10.1016/j.heliyon.2024.e38601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
Objective We conducted a retrospective study on the prognostic factors of aneurysmal subarachnoid hemorrhage (aSAH) patients in the author's Hospital from January 2019 to May 2023. To discuss the association of the blood lipid and glucose levels of patients with the prognosis of aSAH, and verify that high blood lipid and glucose levels are important factors affecting adverse prognosis. Methods All patients with aSAH were collected as the case group, which was divided into two groups according to the modified Rankin Scores (mRS), the good prognosis group (Group A, mRS < 3) and the adverse prognosis group (Group B,mRS ≥3). The clinical data of age, gender, accompanied chronic diseases (hypertension, diabetes), smoking, drinking, Glasgow Coma Scale (GCS), Hunt-Hess (H-H) grade, Modified Fisher grade, total cholesterol (TC) , triglyceride (TG) , high-density cholesterol lipoprotein (HDL-C) , low-density cholesterol lipoprotein (LDL-C) , blood glucose (BG) , responsible aneurysm diameter and location were recorded too. Correlations between blood lipid and glucose levels and Modified Fisher grade were assessed by the Spearman correlation analysis. The receiver operating characteristic (ROC) curve was utilized to evaluate the diagnostic efficacy. The effect of blood lipid and glucose levels on adverse prognosis was analyzed by Logistic regression models. Result A total of 259 patients with aSAH were enrolled. The average age of all patients is (56.54 ± 10.52) years, including 96 males and 163 females. They were divided into Group A (n = 146) and Group B (n = 113). Univariate analysis results show that age, the levels of TC, TG, LDL-C, and BG were higher in Group B (P < 0.05). Besides, Group B had more severe GCS, H-H grade, and Modified Fisher grade than Group A, and a higher proportion of intracranial aneurysms with larger diameter (P < 0.05). Correlation analysis showed that TC, TG, LDL-C, and BG levels were positively correlated with Modified Fisher grade (P < 0.05) and H-H grade (P < 0.05). Multivariate logistic regression model analysis showed that high level of Modified Fisher grade (OR = 0.079, 95%CI: 0.027-0.230) , high level of H-H grade (OR = 0.204, 95%CI: 0.067-0.622) , TC (OR = 10.711, 95%CI: 2.457-46.700) , LDL-C (OR = 0.178, 95%CI: 0.039-0.823) and BG (OR = 1.273, 95%CI: 1.012-1.602) increased the risk of adverse prognosis. The AUC of "H-H grade", "Modified Fisher grade", "TC level", "LDL-C level" and "BG level" was 0.822, 0.885, 0.860, 0.772, and 0.721, respectively, in the ROC curve. Conclusion Modified Fisher grade, H-H grade, TC, LDL-C, and BG levels at admission were independent predictors of adverse prognosis of aSAH. Besides, TC, LDL-C, and BG levels were positively correlated with Modified Fisher grade and Hunt-Hess grade. What's more, high levels of TC, LDL-C, and BG combined with Modified Fisher grade and H-H grade can identify high-risk groups with adverse prognoses in aSAH patients.
Collapse
Affiliation(s)
| | | | - Lei Xingwei
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tang Liuyang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhang Xiaofeng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Li Xiaoguo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guo Zongduo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
3
|
Won SJ, Zhang Y, Butler NJ, Kim K, Mocanu E, Nzoutchoum OT, Lakkaraju R, Davis J, Ghosh S, Swanson RA. Stress hyperglycemia exacerbates inflammatory brain injury after stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594195. [PMID: 38798486 PMCID: PMC11118312 DOI: 10.1101/2024.05.14.594195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Post-stroke hyperglycemia occurs in 30% - 60% of ischemic stroke patients as part of the systemic stress response, but neither clinical evidence nor pre-clinical studies indicate whether post-stroke hyperglycemia affects stroke outcome. Here we investigated this issue using a mouse model of permanent ischemia. Mice were maintained either normoglycemic or hyperglycemic during the interval of 17 - 48 hours after ischemia onset. Post-stroke hyperglycemia was found to increase infarct volume, blood-brain barrier disruption, and hemorrhage formation, and to impair motor recovery. Post-stroke hyperglycemia also increased superoxide formation by peri-infarct microglia/macrophages. In contrast, post-stroke hyperglycemia did not increase superoxide formation or exacerbate motor impairment in p47 phox-/- mice, which cannot form an active superoxide-producing NADPH oxidase-2 complex. These results suggest that hyperglycemia occurring hours-to-days after ischemia can increase oxidative stress in peri-infarct tissues by fueling NADPH oxidase activity in reactive microglia/macrophages, and by this mechanism contribute to worsened functional outcome.
Collapse
|
4
|
Arai-Okuda M, Murai Y, Maeda H, Kanamori A, Miki T, Naito T, Sugihara K, Kono M, Tanito M, Onoe H, Hirooka K, Kiuchi Y, Shinohara M, Kusuhara S, Mori S, Ueda K, Sakamoto M, Yamada-Nakanishi Y, Nakamura M. Potentially compromised systemic and local lactate metabolic balance in glaucoma, which could increase retinal glucose and glutamate concentrations. Sci Rep 2024; 14:3683. [PMID: 38355836 PMCID: PMC10866861 DOI: 10.1038/s41598-024-54383-4] [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: 11/18/2023] [Accepted: 02/12/2024] [Indexed: 02/16/2024] Open
Abstract
To investigate the association between lactate metabolism and glaucoma, we conducted a multi-institutional cross-sectional clinical study and a retinal metabolomic analysis of mice with elevated intraocular pressure (IOP) induced by intracameral microbead injection. We compared lactate concentrations in serum and aqueous humor in age-matched 64 patients each with primary open-angle glaucoma (POAG) and cataract. Neither serum nor aqueous humor lactate concentrations differed between the two groups. Multiple regression analysis revealed that only body mass index showed a significant positive correlation with serum and aqueous humor lactate concentration in POAG patients (rs = 0.376, P = 0.002, and rs = 0.333, P = 0.007, respectively), but not in cataract patients. L-Lactic acid was one of the most abundantly detected metabolites in mouse retinas with gas chromatography and mass spectrometry, but there were no significant differences among control, 2-week, and 4-week IOP elevation groups. After 4 weeks of elevated IOP, D-glucose and L-glutamic acid ranked as the top two for a change in raised concentration, roughly sevenfold and threefold, respectively (ANOVA, P = 0.004; Tukey-Kramer, P < 0.05). Glaucoma may disrupt the systemic and intraocular lactate metabolic homeostasis, with a compensatory rise in glucose and glutamate in the retina.
Collapse
Affiliation(s)
- Mina Arai-Okuda
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yusuke Murai
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | | | - Akiyasu Kanamori
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Kanamori Eye Clinic, Akashi, Japan
| | | | | | - Kazunobu Sugihara
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Michihiro Kono
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Masaki Tanito
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Hiromitsu Onoe
- Department of Ophthalmology and Visual Science, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Kazuyuki Hirooka
- Department of Ophthalmology and Visual Science, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yoshiaki Kiuchi
- Department of Ophthalmology and Visual Science, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Masakazu Shinohara
- Division of Molecular Epidemiology, Department of Future Medical Sciences, Kobe University Graduate School of Medicine, Kobe, Japan
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Sentaro Kusuhara
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Sotaro Mori
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kaori Ueda
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Mari Sakamoto
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yuko Yamada-Nakanishi
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Makoto Nakamura
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
5
|
Ruan Z, Cao G, Qian Y, Fu L, Hu J, Xu T, Wu Y, Lv Y. Single-cell RNA sequencing unveils Lrg1's role in cerebral ischemia‒reperfusion injury by modulating various cells. J Neuroinflammation 2023; 20:285. [PMID: 38037097 PMCID: PMC10687904 DOI: 10.1186/s12974-023-02941-4] [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: 07/28/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Cerebral ischemia‒reperfusion injury causes significant harm to human health and is a major contributor to stroke-related deaths worldwide. Current treatments are limited, and new, more effective prevention and treatment strategies that target multiple cell components are urgently needed. Leucine-rich alpha-2 glycoprotein 1 (Lrg1) appears to be associated with the progression of cerebral ischemia‒reperfusion injury, but the exact mechanism of it is unknown. METHODS Wild-type (WT) and Lrg1 knockout (Lrg1-/-) mice were used to investigate the role of Lrg1 after cerebral ischemia‒reperfusion injury. The effects of Lrg1 knockout on brain infarct volume, blood‒brain barrier permeability, and neurological score (based on 2,3,5-triphenyl tetrazolium chloride, evans blue dye, hematoxylin, and eosin staining) were assessed. Single-cell RNA sequencing (scRNA-seq), immunofluorescence, and microvascular albumin leakage tests were utilized to investigate alterations in various cell components in brain tissue after Lrg1 knockout. RESULTS Lrg1 expression was increased in various cell types of brain tissue after cerebral ischemia‒reperfusion injury. Lrg1 knockout reduced cerebral edema and infarct size and improved neurological function after cerebral ischemia‒reperfusion injury. Single-cell RNA sequencing analysis of WT and Lrg1-/- mouse brain tissues after cerebral ischemia‒reperfusion injury revealed that Lrg1 knockout enhances blood‒brain barrier (BBB) by upregulating claudin 11, integrin β5, protocadherin 9, and annexin A2. Lrg1 knockout also promoted an anti-inflammatory and tissue-repairing phenotype in microglia and macrophages while reducing neuron and oligodendrocyte cell death. CONCLUSIONS Our results has shown that Lrg1 mediates numerous pathological processes involved in cerebral ischemia‒reperfusion injury by altering the functional states of various cell types, thereby rendering it a promising therapeutic target for cerebral ischemia‒reperfusion injury.
Collapse
Affiliation(s)
- Zhaohui Ruan
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guosheng Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yisong Qian
- School of Clinical Medicine, Nanchang University, Nanchang, China
| | - Longsheng Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jinfang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tiantian Xu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yaoqi Wu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanni Lv
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
6
|
Shaheryar ZA, Khan MA, Hameed H, Zaidi SAA, Anjum I, Rahman MSU. Lauric acid provides neuroprotection against oxidative stress in mouse model of hyperglycaemic stroke. Eur J Pharmacol 2023; 956:175990. [PMID: 37572940 DOI: 10.1016/j.ejphar.2023.175990] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/14/2023]
Abstract
During ischemic stroke, higher glucose level linked worse outcomes were reported even in patients without pre-existing diabetes. Evidence suggest that such worse stroke outcomes were mainly due to production of reactive, toxic glucose metabolites that expands oxidative damage inside the brain. As a consequence of high oxidative stress, microvasculature structures and tight junctions compromised their functionally, infarct volume expands and brain edema exacerbates. In a mouse model of ischemic stroke with induced acute hyperglycaemia, Lauric acid (LA) as a natural saturated fatty acid demonstrated neuroprotection by attenuating infarct volume and brain edema. In addition, in the ipsilateral hyperglycaemic brain, the LA significantly increased the expression of tight junction representative protein (occludin) as well as anti-oxidative markers; Manganese superoxide dismutase (Mn) SOD, Extracellular superoxide dismutase (Ec-SOD) and nuclear factor-erythroid factor 2-related factor 2 (Nrf2) in the ipsilateral region against hyperglycemic ischemic stroke. LA treated animals showed a significant reduction in the production of lipid peroxidation products (4-HNE) in the microvascular structures, maintained the blood brain barrier (BBB) integrity. LA linked neuroprotective outcomes were further confirmed by behavioral tests, where functional outcomes and motor coordination were improved significantly. Furthermore, LA treatment enhanced food intake, decreased mortality rate, and net body weight loss. Conclusively, LA modulated ischemic insult exacerbated by hyperglycemia and provided neuroprotection.
Collapse
Affiliation(s)
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Syed Awais Ali Zaidi
- Department of Pharmacy, The Sahara University Narowal, Narowal, 51600, Pakistan.
| | - Irfan Anjum
- Faculty of Pharmacy, The University of Lahore, Lahore, 54000, Pakistan; Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-E-Milat University, Islamabad, Pakistan.
| | | |
Collapse
|
7
|
Zhang H, Du D, Gao X, Tian X, Xu Y, Wang B, Yang S, Liu P, Li Z. PFT-α protects the blood-brain barrier through the Wnt/β-catenin pathway after acute ischemic stroke. Funct Integr Genomics 2023; 23:314. [PMID: 37777676 DOI: 10.1007/s10142-023-01237-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
The dysfunction of blood-brain barrier (BBB) plays a pivotal role in brain injury and subsequent neurological deficits of ischemic stroke. The current study aimed to examine the potential correlation between p53 inhibition and the neuroprotective effect of on the BBB. Rat middle cerebral artery occlusion and reperfusion model (MCAO/R) and oxygen-glucose deprivation/re-oxygenation model (OGD/R) were employed to simulate cerebral ischemia-reperfusion (CI/R) injury occurrence in vivo and in vitro. mNSS and TTC staining were applied to evaluate neurological deficits and brain infarct volumes. Evans blue (EB) staining was carried out to examine the permeability of BBB. RT-qPCR and Western blot to examine the mRNA and protein levels. Cell viabilities were detected by CCK-8. Flow cytometry and ELISA assay were employed to examine apoptosis and neuroinflammation levels. TEER value and sodium fluorescein were carried out to explore the permeability of HBMEC cells. PFT-α inhibited P53 and promoted the expression of β-catenin and cyclin D1, which were reversed by DKK1. PFT-α inhibited neurological deficits, brain infarct volume, neuroinflammation, apoptosis, and BBB integrity than the MCAO/R rats; however, this inhibition was reversed by DKK1. PFT-α promoted OGD/R-induced cell viability in NSCs, and suppressed inflammation and apoptosis, but DKK1 weakened the effect of PFT-α. PFT-α increased OGD/R-induced TEER values in cerebrovascular endothelial cells, inhibited sodium fluorescein permeability, and increased the mRNA levels of tight junction protein, but they were all attenuated by DKK1. PFT-α protects the BBB after acute ischemic stroke via the Wnt/β-catenin pathway, which in turn improves neurological function.
Collapse
Affiliation(s)
- Haitao Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Deyong Du
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Xiaoning Gao
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Xiaoling Tian
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Yongqiang Xu
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Bo Wang
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China
| | - Shoujuan Yang
- Department of Cardiology, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China.
| | - Pengfei Liu
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China.
| | - Zefu Li
- Department of Neurosurgery, Binzhou Medical University Hospital, No. 661, Huanghe 2nd Road, Binzhou, 256603, China.
| |
Collapse
|
8
|
Mannismäki L, Martinez-Majander N, Sibolt G, Suomalainen OP, Bäcklund K, Abou Elseoud A, Järveläinen J, Forss N, Curtze S. Association of admission plasma glucose level and cerebral computed tomographic perfusion deficit volumes. J Neurol Sci 2023; 451:120722. [PMID: 37393736 DOI: 10.1016/j.jns.2023.120722] [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: 04/12/2023] [Revised: 06/19/2023] [Accepted: 06/24/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Hyperglycemia in acute ischemic stroke (AIS) is frequent and associated with worse outcome. Yet, strict glycemic control in AIS patients has failed to yield beneficial outcome. So far, the underlying pathophysiological mechanisms of admission hyperglycemia in AIS have remained not fully understood. We aimed to evaluate the yet equivocal association of hyperglycemia with computed tomographic perfusion (CTP) deficit volumes. PATIENTS AND METHODS We included 832 consecutive AIS and transient ischemic attack (TIA) patients who underwent CTP as a part of screening for recanalization treatment (stroke code) between 3/2018 and 10/2020, from the prospective cohort of Helsinki Stroke Quality Registry. Associations of admission glucose level (AGL) and CTP deficit volumes, namely ischemic core, defined as relative cerebral blood flow <30%, and hypoperfusion lesions Time-to-maximum (Tmax) >6 s and Tmax >10s, as determined with RAPID® software, were analyzed with a linear regression model adjusted for age, sex, C-reactive protein, and time from symptom onset to imaging. RESULTS AGL median was 6.8 mmol/L (interquartile range 5.9-8.0 mmol/L), and 222 (27%) patients were hyperglycemic (glucose >7.8 mmol/L) on admission. In non-diabetic patients (643 [77%]), AGL was significantly associated with volume of Tmax. >6 s (regression coefficient [RC] 4.8, 95% confidence interval [CI] 0.49-9.1), of Tmax >10s (RC 4.6, 95% CI 1.2-8.1), and of ischemic core (RC 2.6, 95% CI 0.64-4.6). No significant associations were shown in diabetic patients. CONCLUSION Admission hyperglycemia appears to be associated with both larger volume of hypoperfusion lesions and of ischemic core in non-diabetic stroke code patients with AIS and TIA.
Collapse
Affiliation(s)
- Laura Mannismäki
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, Helsinki, Finland.
| | - Nicolas Martinez-Majander
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Gerli Sibolt
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Olli P Suomalainen
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Katariina Bäcklund
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Ahmed Abou Elseoud
- Helsinki Medical Imaging Centre, Helsinki University Hospital, Helsinki, Finland
| | - Juha Järveläinen
- Helsinki Medical Imaging Centre, Helsinki University Hospital, Helsinki, Finland
| | - Nina Forss
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, Helsinki, Finland; Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Sami Curtze
- Department of Neurology, Helsinki University Hospital and Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Kittur FS, Hung CY, Li PA, Sane DC, Xie J. Asialo-rhuEPO as a Potential Neuroprotectant for Ischemic Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:610. [PMID: 37111367 PMCID: PMC10143832 DOI: 10.3390/ph16040610] [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: 03/28/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Neuroprotective drugs to protect the brain against cerebral ischemia and reperfusion (I/R) injury are urgently needed. Mammalian cell-produced recombinant human erythropoietin (rhuEPOM) has been demonstrated to have excellent neuroprotective functions in preclinical studies, but its neuroprotective properties could not be consistently translated in clinical trials. The clinical failure of rhuEPOM was thought to be mainly due to its erythropoietic activity-associated side effects. To exploit its tissue-protective property, various EPO derivatives with tissue-protective function only have been developed. Among them, asialo-rhuEPO, lacking terminal sialic acid residues, was shown to be neuroprotective but non-erythropoietic. Asialo-rhuEPO can be prepared by enzymatic removal of sialic acid residues from rhuEPOM (asialo-rhuEPOE) or by expressing human EPO gene in glycoengineered transgenic plants (asialo-rhuEPOP). Both types of asialo-rhuEPO, like rhuEPOM, displayed excellent neuroprotective effects by regulating multiple cellular pathways in cerebral I/R animal models. In this review, we describe the structure and properties of EPO and asialo-rhuEPO, summarize the progress on neuroprotective studies of asialo-rhuEPO and rhuEPOM, discuss potential reasons for the clinical failure of rhuEPOM with acute ischemic stroke patients, and advocate future studies needed to develop asialo-rhuEPO as a multimodal neuroprotectant for ischemic stroke treatment.
Collapse
Affiliation(s)
- Farooqahmed S. Kittur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - Chiu-Yueh Hung
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - David C. Sane
- Carilion Clinic and Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA;
| | - Jiahua Xie
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| |
Collapse
|
10
|
The Prognostic Significance of Early Glycemic Profile in Acute Ischemic Stroke Depends on Stroke Subtype. J Clin Med 2023; 12:jcm12051794. [PMID: 36902581 PMCID: PMC10003561 DOI: 10.3390/jcm12051794] [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: 01/25/2023] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023] Open
Abstract
It is still unclear whether early glycemic profile after admission for acute ischemic stroke (IS) has the same prognostic significance in patients with lacunar and non-lacunar infarction. Data from 4011 IS patients admitted to a Stroke Unit (SU) were retrospectively analyzed. Lacunar IS was diagnosed by clinical criteria. A continuous indicator of early glycemic profile was calculated as the difference of fasting serum glucose (FSG) measured within 48 h after admission and random serum glucose (RSG) measured on admission. Logistic regression was used to estimate the association with a combined poor outcome defined as early neurological deterioration, severe stroke at SU discharge, or 1-month mortality. Among patients without hypoglycemia (RSG and FSG > 3.9 mmol/L), an increasing glycemic profile increased the likelihood of a poor outcome for non-lacunar (OR, 1.38, 95%CI, 1.24-1.52 in those without diabetes; 1.11, 95%CI, 1.05-1.18 in those with diabetes) but not for lacunar IS. Among patients without sustained or delayed hyperglycemia (FSG < 7.8 mmol/L), an increasing glycemic profile was unrelated to outcome for non-lacunar IS but decreased the likelihood of poor outcome for lacunar IS (OR, 0.63, 95%CI, 0.41-0.98). Early glycemic profile after acute IS has a different prognostic significance in non-lacunar and lacunar patients.
Collapse
|
11
|
Lyu N, Li X. Sevoflurane Postconditioning Attenuates Cerebral Ischemia-Reperfusion Injury by Inhibiting SP1/ACSL4-Mediated Ferroptosis. Hum Exp Toxicol 2023; 42:9603271231160477. [PMID: 36842993 DOI: 10.1177/09603271231160477] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Sevoflurane is the most commonly used anesthetic in clinical practice and exerts a protective effect on cerebral ischemia-reperfusion (I/R) injury. This study aims to elucidate the molecular mechanism by which sevoflurane postconditioning protects against cerebral I/R injury. Oxygen-glucose deprivation/reperfusion (OGD/R) model in vitro and the middle cerebral artery occlusion (MCAO) model in vivo were established to simulate cerebral I/R injury. Sevoflurane postconditioning reduced neurological deficits, cerebral infarction, and ferroptosis after I/R injury. Interestingly, sevoflurane significantly inhibited specificity protein 1 (SP1) expression in MACO rats and HT22 cells exposed to OGD/R. SP1 overexpression attenuated the neuroprotective effects of sevoflurane on OGD/R-treated HT22 cells, evidenced by reduced cell viability, increased apoptosis, and cleaved caspase-3 expression. Furthermore, chromatin immunoprecipitation and luciferase experiments verified that SP1 bound directly to the ACSL4 promoter region to increase its expression. In addition, sevoflurane inhibited ferroptosis via SP1/ACSL4 axis. Generally, our study describes an anti-ferroptosis effect of sevoflurane against cerebral I/R injury via downregulating the SP1/ASCL4 axis. These findings suggest a novel sight for cerebral protection against cerebral I/R injury and indicate a potential therapeutic approach for a variety of cerebral diseases.
Collapse
Affiliation(s)
- Ning Lyu
- Department of Anesthesiology, 56659Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Xiaoyun Li
- Department of Anesthesiology, 144991The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
12
|
Temporal Pattern of Neuroinflammation Associated with a Low Glycemic Index Diet in the 5xFAD Mouse Model of Alzheimer's Disease. Mol Neurobiol 2022; 59:7303-7322. [PMID: 36175825 PMCID: PMC9616770 DOI: 10.1007/s12035-022-03047-3] [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: 04/26/2022] [Accepted: 09/21/2022] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) is associated with brain amyloid-β (Aβ) peptide accumulation and neuroinflammation. Currants, a low glycemic index dried fruit, and their components display pleiotropic neuroprotective effects in AD. We examined how diet containing 5% Corinthian currant paste (CurD) administered in 1-month-old 5xFAD mice for 1, 3, and 6 months affects Aβ levels and neuroinflammation in comparison to control diet (ConD) or sugar-matched diet containing 3.5% glucose/fructose (GFD). No change in serum glucose or insulin levels was observed among the three groups. CurD administered for 3 months reduced brain Aβ42 levels in male mice as compared to ConD and GFD, but after 6 months, Aβ42 levels were increased in mice both on CurD and GFD compared to ConD. CurD for 3 months also reduced TNFα and IL-1β levels in male and female mouse cortex homogenates compared to ConD and GFD. However, after 6 months, TNFα levels were increased in cortex homogenates of mice both on CurD and GFD as compared to ConD. A similar pattern was observed for TNFα-expressing cells, mostly co-expressing the microglial marker CD11b, in mouse hippocampus. IL-1β levels were similarly increased in the brain of all groups after 6 months. Furthermore, a time dependent decrease of secreted TNFα levels was found in BV2 microglial cells treated with currant phenolic extract as compared to glucose/fructose solution. Overall, our findings suggest that a short-term currant consumption reduces neuroinflammation in 5xFAD mice as compared to sugar-matched or control diet, but longer-term intake of currant or sugar-matched diet enhances neuroinflammation.
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Outcome following traumatic brain injury (TBI) remains variable, and derangements in cerebral metabolism are a common finding in patients with poor outcome. This review compares our understanding of cerebral metabolism in health with derangements seen following TBI. RECENT FINDINGS Ischemia is common within the first 24 h of injury and inconsistently detected by bedside monitoring. Metabolic derangements can also result from tissue hypoxia in the absence of ischemic reductions in blood flow due to microvascular ischemia and mitochondrial dysfunction. Glucose delivery across the injured brain is dependent on blood glucose and regional cerebral blood flow, and is an important contributor to derangements in glucose metabolism. Alternative energy substrates such as lactate, ketone bodies and succinate that may support mitochondrial function, and can be utilized when glucose availability is low, have been studied following TBI but require further investigation. SUMMARY Mitochondrial dysfunction and the use of alternative energy substrates are potential therapeutic targets, but improved understanding of the causes, impact and significance of metabolic derangements in clinical TBI are needed. Maintaining adequate oxygen and glucose delivery across the injured brain may accelerate the recovery of mitochondrial function and cerebral energy metabolism and remain important management targets.
Collapse
Affiliation(s)
- Simon Demers-Marcil
- Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- Department of Anesthesiology and Critical Care, CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Jonathan P. Coles
- Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
14
|
Guo S, Wehbe A, Syed S, Wills M, Guan L, Lv S, Li F, Geng X, Ding Y. Cerebral Glucose Metabolism and Potential Effects on Endoplasmic Reticulum Stress in Stroke. Aging Dis 2022; 14:450-467. [PMID: 37008060 PMCID: PMC10017147 DOI: 10.14336/ad.2022.0905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Ischemic stroke is an extremely common pathology with strikingly high morbidity and mortality rates. The endoplasmic reticulum (ER) is the primary organelle responsible for conducting protein synthesis and trafficking as well as preserving intracellular Ca2+ homeostasis. Mounting evidence shows that ER stress contributes to stroke pathophysiology. Moreover, insufficient circulation to the brain after stroke causes suppression of ATP production. Glucose metabolism disorder is an important pathological process after stroke. Here, we discuss the relationship between ER stress and stroke and treatment and intervention of ER stress after stroke. We also discuss the role of glucose metabolism, particularly glycolysis and gluconeogenesis, post-stroke. Based on recent studies, we speculate about the potential relationship and crosstalk between glucose metabolism and ER stress. In conclusion, we describe ER stress, glycolysis, and gluconeogenesis in the context of stroke and explore how the interplay between ER stress and glucose metabolism contributes to the pathophysiology of stroke.
Collapse
Affiliation(s)
- Sichao Guo
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Alexandra Wehbe
- Department of Neurosurgery, Wayne State University School of Medicine, USA
- Harvard T.H. Chan School of Public Health, USA
| | - Shabber Syed
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Melissa Wills
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Longfei Guan
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Shuyu Lv
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
- Correspondence should be addressed to: Dr. Xiaokun Geng, Beijing Luhe Hospital, Capital Medical University, Beijing, China. E-mail: ; Dr. Yuchuan Ding, Wayne State University School of Medicine, Detroit, MI 48201, USA. E-mail:
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, USA
- Correspondence should be addressed to: Dr. Xiaokun Geng, Beijing Luhe Hospital, Capital Medical University, Beijing, China. E-mail: ; Dr. Yuchuan Ding, Wayne State University School of Medicine, Detroit, MI 48201, USA. E-mail:
| |
Collapse
|