51
|
Chu H, Dong J, Tang Y, Huang C, Guo Q. Connexin 43 Promotes Neurogenesis via Regulating Aquaporin-4 after Cerebral Ischemia. Neurotox Res 2023; 41:349-361. [PMID: 37074591 DOI: 10.1007/s12640-023-00646-3] [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: 01/25/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 04/20/2023]
Abstract
We aimed to test the effects of connexin43 (Cx43) on ischemic neurogenesis and examined whether it was dependent on aquaporin-4 (AQP4). We detected the expression of Cx43 and AQP4 in the ipsilateral subventricular zone (SVZ) and peri-infarct cortex after middle cerebral artery occlusion (MCAO). Also, we examined neurogenesis in the above regions via co-labeling of 5-bromo-2-deoxyuridine (BrdU)/neuronal nuclear antigen (NeuN) and BrdU/doublecortin (DCX). The effects of Cx43 and AQP4 were investigated by using two transgenic animals: heterozygous Cx43 (Cx43±) mice and AQP4 knockout (AQP4-/-) mice, and connexin mimetic peptide (CMP), a selective Cx43 blocker. We demonstrated AQP4 and Cx43 were co-expressed in the astrocytes after MCAO and the expression was highly increased in ipsilateral SVZ and peri-infarct cortex. Cx43± mice had larger infarction volumes and worse neurological function. Both BrdU/NeuN and BrdU/DCX co-labeled cells in the two regions were reduced in Cx43± and AQP4-/- mice compared to wild-type (WT) mice, suggesting Cx43 and AQP4 participated in neurogenesis of neural stem cells. Moreover, CMP decreased AQP4 expression and inhibited neurogenesis in WT mice, while the latter failed to be observed in AQP4-/- mice. Besides, higher levels of IL-1β and TNF-α were detected in the SVZ and peri-infarct cortex of AQP4-/- and Cx43± mice than those in WT mice. In conclusion, our data suggest that Cx43 elicits neuroprotective effects after cerebral ischemia through promoting neurogenesis in the SVZ to regenerate the injured neurons, which is AQP4 dependent and associated with down-regulation of inflammatory cytokines IL-1β and TNF-α.
Collapse
Affiliation(s)
- Heling Chu
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, 200233, Shanghai, China
| | - Jing Dong
- Department of Internal Neurology, Qingdao Municipal Hospital, Qingdao, China
| | - Yuping Tang
- Department of Neurology, Huashan Hospital, Fudan University, No. 12 Mid. Wulumuqi Road, Shanghai, 200040, China.
| | - Chuyi Huang
- Health Management Center, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai, 200120, China.
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, 200233, Shanghai, China.
| |
Collapse
|
52
|
Tai SH, Chao LC, Huang SY, Lin HW, Lee AH, Chen YY, Lee EJ. Nicotinamide Deteriorates Post-Stroke Immunodepression Following Cerebral Ischemia-Reperfusion Injury in Mice. Biomedicines 2023; 11:2145. [PMID: 37626642 PMCID: PMC10452067 DOI: 10.3390/biomedicines11082145] [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: 06/20/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Inducing experimental stroke leads to biphasic immune responses, where the early activation of immune functions is followed by severe immunosuppression accompanied by spleen and thymus atrophy. Nicotinamide, a water-soluble B-group vitamin, is a known neuroprotectant against brain ischemia in animal models. We examined the effect of nicotinamide on the central and peripheral immune response in experimental stroke models. (2) Methods: Nicotinamide (500 mg/kg) or saline was intravenously administered to C57BL/6 mice during reperfusion after transiently occluding the middle cerebral artery or after LPS injection. On day 3, the animals were examined for behavioral performance and were then sacrificed to assess brain infarction, blood-brain barrier (BBB) integrity, and the composition of immune cells in the brain, thymus, spleen, and blood using flow cytometry. (3) Results: Nicotinamide reduced brain infarction and microglia/macrophage activation following MCAo (p < 0.05). Similarly, in LPS-injected mice, microglia/macrophage activation was decreased upon treatment with nicotinamide (p < 0.05), suggesting a direct inhibitory effect of nicotinamide on microglia/macrophage activation. Nicotinamide decreased the infiltration of neutrophils into the brain parenchyma and ameliorated Evans blue leakage (p < 0.05), suggesting that a decreased infiltration of neutrophils could, at least partially, be the result of a more integrated BBB structure following nicotinamide treatment. Our studies also revealed that administering nicotinamide led to retarded B-cell maturation in the spleen and subsequently decreased circulating B cells in the thymus and bloodstream (p < 0.05). (4) Conclusions: Cumulatively, nicotinamide decreased brain inflammation caused by ischemia-reperfusion injury, which was mediated by a direct anti-inflammatory effect of nicotinamide and an indirect protective effect on BBB integrity. Administering nicotinamide following brain ischemia resulted in a decrease in circulating B cells. This warrants attention with respect to future clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - E-Jian Lee
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| |
Collapse
|
53
|
Qin YY, Pan SY, Dai JR, Wang QM, Luo X, Qin ZH, Luo L. Alleviation of ischemic brain injury by exercise preconditioning is associated with modulation of autophagy and mitochondrial dynamics in cerebral cortex of female aged mice. Exp Gerontol 2023; 178:112226. [PMID: 37257699 DOI: 10.1016/j.exger.2023.112226] [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: 02/24/2023] [Revised: 05/17/2023] [Accepted: 05/27/2023] [Indexed: 06/02/2023]
Abstract
Evidence from clinical studies and preclinical studies supports that exercise preconditioning can not only reduce the risk of stroke but also improve brain tissue and functional outcome after stroke. It has been demonstrated that autophagy and mitochondrial dynamics are involved in ischemic stroke. However, it is still unclear whether exercise preconditioning-induced neuroprotection against stroke is associated with modulation of autophagy and mitochondrial dynamics. Although age and sex interactively affect ischemic stroke risk, incidence, and outcome, studies based on young male animals are most often used to explore the role of exercise preconditioning in the prevention of ischemic stroke. In the current study, we examined whether exercise preconditioning could modulate autophagy and mitochondrial dynamics in a brain ischemia and reperfusion (I/R) model of female aged mice. The results showed that exercise preconditioning reduced infarct volume and improved neurological deficits. Additionally, increased levels of autophagy-related proteins LC3-II/LC3-I, LC3-II, p62, Atg7, and mitophagy-related proteins Bnip3L and Parkin, as well as increased levels of mitochondrial fusion modulator Mfn2 and mitochondrial fission modulator Drp1 in the ischemic cortex of female aged mice at 12 h after I/R were present. Our results could contribute to a better understanding of exercise preconditioning-induced neuroprotection against ischemic stroke for the elderly.
Collapse
Affiliation(s)
- Yuan-Yuan Qin
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215009, Jiangsu Province, China; Department of Pharmacy, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, Jiangsu 215009, China
| | - Shan-Yao Pan
- School of Physical Education and Sports Science, Soochow University; Suzhou 215021, China
| | - Jia-Ru Dai
- School of Physical Education and Sports Science, Soochow University; Suzhou 215021, China
| | - Qing-Mei Wang
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Teaching Affiliate of Harvard Medical School, Charlestown, MA, USA
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases (SZS0703); Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Soochow University School of Pharmaceutical Science; Suzhou 215123, China
| | - Li Luo
- School of Physical Education and Sports Science, Soochow University; Suzhou 215021, China.
| |
Collapse
|
54
|
Li Y, Tan L, Yang C, He L, Liu L, Deng B, Liu S, Guo J. Distinctions between the Koizumi and Zea Longa methods for middle cerebral artery occlusion (MCAO) model: a systematic review and meta-analysis of rodent data. Sci Rep 2023; 13:10247. [PMID: 37353569 PMCID: PMC10290095 DOI: 10.1038/s41598-023-37187-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/17/2023] [Indexed: 06/25/2023] Open
Abstract
Ischemic stroke in rodents is usually induced by intraluminal middle cerebral artery occlusion (MCAO) via the common carotid artery plugging filament invented by Koizumi et al. (MCAO-KM), or the external carotid artery plugging filament created by Zea Longa et al. (MCAO-LG). A systematic review of the distinctions between them is currently lacking. Here, we performed a meta-analysis in terms of model establishment, cerebral blood flow (CBF), and cerebral ischemia-reperfusion injury (CIRI) between them, Weighted Mean Differences and Standardized Mean Difference were used to analyze the combined effects, Cochrane's Q test and the I2 statistic were applied to determine heterogeneity, sensitivity analysis and subgroup analysis were performed to explore the source of heterogeneity. Literature mining suggests that MCAO-KM brings shorter operation time (p = 0.007), higher probability of plugging filament (p < 0.001) and molding establishment (p = 0.006), lower possibility of subarachnoid hemorrhage (p = 0.02), larger infarct volume (p = 0.003), severer brain edema (p = 0.002), and neurological deficits (p = 0.03). Nevertheless, MCAO-LG shows a more adequate CBF after ischemia-reperfusion (p < 0.001), a higher model survival rate (p = 0.02), and a greater infarct rate (p = 0.007). In conclusion, the MCAO-KM method is simple to operate with a high modeling success rate, and is suitable for the study of brain edema under long-term hypoperfusion, while the MCAO-LG method is highly challenging for novices, and is suitable for the study of CIRI caused by complete ischemia-reperfusion. These findings are expected to benefit the selection of intraluminal filament MCAO models before undertaking ischemic stroke preclinical effectiveness trials.
Collapse
Affiliation(s)
- Yong Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Tan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Caixia Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liying He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bowen Deng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sijing Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlin Guo
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
55
|
Balkaya M, Dohare P, Chen S, Schober AL, Fidaleo AM, Nalwalk JW, Sah R, Mongin AA. Conditional deletion of LRRC8A in the brain reduces stroke damage independently of swelling-activated glutamate release. iScience 2023; 26:106669. [PMID: 37182109 PMCID: PMC10173736 DOI: 10.1016/j.isci.2023.106669] [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: 12/30/2022] [Revised: 03/03/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
The ubiquitous volume-regulated anion channels (VRACs) facilitate cell volume control and contribute to many other physiological processes. Treatment with non-specific VRAC blockers or brain-specific deletion of the essential VRAC subunit LRRC8A is highly protective in rodent models of stroke. Here, we tested the widely accepted idea that the harmful effects of VRACs are mediated by release of the excitatory neurotransmitter glutamate. We produced conditional LRRC8A knockout either exclusively in astrocytes or in the majority of brain cells. Genetically modified mice were subjected to an experimental stroke (middle cerebral artery occlusion). The astrocytic LRRC8A knockout yielded no protection. Conversely, the brain-wide LRRC8A deletion strongly reduced cerebral infarction in both heterozygous (Het) and full KO mice. Yet, despite identical protection, Het mice had full swelling-activated glutamate release, whereas KO animals showed its virtual absence. These findings suggest that LRRC8A contributes to ischemic brain injury via a mechanism other than VRAC-mediated glutamate release.
Collapse
Affiliation(s)
- Mustafa Balkaya
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Preeti Dohare
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Sophie Chen
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Alexandra L. Schober
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Antonio M. Fidaleo
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Julia W. Nalwalk
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Rajan Sah
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander A. Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
- Corresponding author
| |
Collapse
|
56
|
Kumari R, Kareem ZY, McLaughlin PJ. Acute Low Dose Naltrexone Increases β-Endorphin and Promotes Neuronal Recovery Following Hypoxia-Ischemic Stroke in Type-2 Diabetic Mice. Neurochem Res 2023:10.1007/s11064-023-03938-4. [PMID: 37166576 DOI: 10.1007/s11064-023-03938-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/05/2023] [Accepted: 04/08/2023] [Indexed: 05/12/2023]
Abstract
Diabetic patients experience significant mortality and poor recovery following ischemic stroke. Our clinical and basic science studies demonstrate an overall immune suppression in the periphery of diabetic stroke patients, as well as within the central nervous system (CNS) of type-2 diabetic mice following hypoxia-ischemia (HI). Low doses of naltrexone (LDN) improved clinical outcomes in many autoimmune diseases by acting on opioid receptors to release β-endorphin which in turn balances inflammatory cytokines and modulates the opioid growth factor (OGF)-opioid growth factor receptor (OGFr) pathway. We hypothesized that in our model of diabetic mice, LDN treatment will induce the release of β-endorphin and improve CNS response by promoting neuronal recovery post HI. To test this hypothesis, we induced HI in 10 week old male db/db and db/ + mice, collected tissue at 24 and 72 h post HI, and measured OGF levels in plasma and brain tissue. The infarct size and number of OGF + neurons in the motor cortex, caudate and hippocampus (CA3) were measured. Following HI, db/db mice had significant increases in brain OGF expression, increased infarct size and neurological deficits, and loss of OGFr + neurons in several different brain regions. In the second experiment, we injected LDN (1 mg/kg) intraperitoneally into db/db and db/ + mice at 4, 24, and 48 h post HI, and collected brain tissue and blood at 72 h. Acute LDN treatment increased β-endorphin and OGF levels in plasma and promoted neuronal recovery in db/db mice compared to phosphate buffer saline (PBS)-treated diabetic mice suggesting a protective or regenerative effect of LDN.
Collapse
Affiliation(s)
- Rashmi Kumari
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Drive H109, Hershey, PA, 17033, USA.
| | - Zainab Y Kareem
- Kent State University College of Podiatric Medicine, 6000 Rockside Woods Boulevard North, Independence, OH, 44131, USA
| | - Patricia J McLaughlin
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Drive H109, Hershey, PA, 17033, USA
| |
Collapse
|
57
|
Nguyen LTT, Le XT, Pham HNT, Van Nguyen T, Nguyen PT, Van Thi Pham A, Nguyen TBT, Matsumoto K. Therapeutic effects of a standardized-flavonoid Diospyros kaki L.f. leaf extract on transient focal cerebral ischemia-induced brain injury in mice. J Nat Med 2023; 77:544-560. [PMID: 37115470 DOI: 10.1007/s11418-023-01699-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023]
Abstract
This study aimed to investigate the neuroprotective and therapeutic effects of Diospyros kaki L.f. leaves (DK) on transient focal cerebral ischemic injury and underlying mechanisms using a middle cerebral artery occlusion (MCAO) model of mice. The animals received the MCAO operation on day 0. The daily administrations of DK (50 and 100 mg/kg, p.o) and edaravone (6 mg/kg, i.v), a reference drug with radical scavenging activity, were started 7 days before (pre-treatment) or immediately after the MCAO operation (post-treatment) and continued during the experimental period. Histochemical, biochemical, and neurological changes and cognitive performance were evaluated. MCAO caused cerebral infarction and neuronal cell loss in the cortex, striatum, and hippocampus in a manner accompanied by spatial cognitive deficits. These neurological and cognitive impairments caused by MCAO were significantly attenuated by pre- and post-ischemic treatments with DK and edaravone, suggesting that DK, like edaravone, has therapeutic potential for cerebral ischemia-induced brain damage. DK and edaravone suppressed MCAO-induced changes in biomarkers for apoptosis (TUNEL-positive cell number and cleaved caspase-3 protein expression) and oxidative stress (glutathione and malondialdehyde contents) in the brain. Interestingly, DK, but not edaravone, mitigated an increase in blood-brain permeability and down-regulation of vascular endothelial growth factor protein expression caused by MCAO. Although the exact chemical constituents implicated in the effects of DK remain to be clarified, the present results indicate that DK exerts neuroprotective and therapeutic activity against transient focal cerebral ischemia-induced injury probably by suppressing oxidative stress, apoptotic process, and mechanisms impairing blood-brain barrier integrity in the brain.
Collapse
Affiliation(s)
- Loan Thanh Thi Nguyen
- Department of Pharmacology and Biochemistry, National Institute of Medicinal Materials, Hanoi, 10000, Vietnam
- Department of Pharmacology, Hanoi Medical University, Hanoi, 10000, Vietnam
| | - Xoan Thi Le
- Department of Pharmacology and Biochemistry, National Institute of Medicinal Materials, Hanoi, 10000, Vietnam.
| | - Hang Nguyet Thi Pham
- Department of Pharmacology and Biochemistry, National Institute of Medicinal Materials, Hanoi, 10000, Vietnam
| | - Tai Van Nguyen
- Department of Phytochemistry, National Institute of Medicinal Materials, Hanoi, 10000, Vietnam
| | - Phuong Thi Nguyen
- Department of Pharmacology and Biochemistry, National Institute of Medicinal Materials, Hanoi, 10000, Vietnam
| | - Anh Van Thi Pham
- Department of Pharmacology, Hanoi Medical University, Hanoi, 10000, Vietnam
| | | | - Kinzo Matsumoto
- Graduate School of Pharmaceutical Sciences, Daiichi University of Pharmacy, Fukuoka, 815-8511, Japan
| |
Collapse
|
58
|
Liu JF, Su G, Chen LX, Zhou JP, Gao J, Zhang JJ, Wu QH, Chen W, Chen DY, Zhang ZC. Irisin Attenuates Apoptosis Following Ischemia-Reperfusion Injury Through Improved Mitochondria Dynamics and ROS Suppression Mediated Through the PI3K/Akt/mTOR Axis. Mol Neurobiol 2023:10.1007/s12035-023-03336-5. [PMID: 37060502 DOI: 10.1007/s12035-023-03336-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 04/03/2023] [Indexed: 04/16/2023]
Abstract
Irisin is a muscle-derived hormone that promotes the survival of motor neurons and enhances muscle size following injury. In this study, we investigated the beneficial effects and mechanism(s) of action of irisin in response to cerebral ischemia-reperfusion injury (CIRI). Right-middle cerebral artery occlusion (MCAO) and hypoxia/reoxygenation (H/R) models were generated in C57BL/6 J mice. Mouse neuronal cell lines (NSC-34) were used to confirm the molecular mechanisms of the protection afforded by irisin in response to CIRI. We found that irisin (250 μg/kg) improved cerebral function and reduced the cerebral infarct volume following CIRI. Irisin also protected neuronal cells against ischemia-reperfusion (I/R) induced apoptosis, assessed via TUNEL, and cleaved Caspase-3 staining. Western blotting of neuronal tissue from irisin treated I/R mice showed lower expression of pro-apoptotic Bax and caspase-9 (P < 0.001 and P < 0.01) and increased levels of the pro-survival protein Bcl-2 (P < 0.01 & P < 0.001 vs. I/R). Irisin also reduced the levels of reactive oxygen species (ROS) characterized through malondialdehyde (MDA) assays. Irisin was found to maintain mitochondrial homeostasis through the suppression of mitochondrial fission-linked dynamin-related protein 1 in CIRI mice (P < 0.01 and P < 0.05 v. I/R cohort). Moreover, mitochondrial fusion-related protein (Mfn2) and Opa1 expression were rescued following irisin treatment (P < 0.001 and P < 0.01 v. I/R cohort). Cell-based assays showed that irisin activates PI3K/AKT/mTOR signaling in the neurons of CIRI mice. Furthermore, the beneficial effects of irisin on NSC-34 cell-survival, mitochondrial function, and ROS generation were reversed by VS-5584, a highly specific PI3K/AKT/mTOR inhibitor. Collectively, these data highlight the ability of irisin to alleviate CIRI in vivo and in vitro. The mechanisms of action of irisin include the attenuation of apoptosis through the prevention of mitochondrial fission and increased mitochondrial fusion and the alleviation of oxidative stress through activation of the PI3K/AKT/mTOR axis. We therefore identify irisin as a much-needed therapeutic for CIRI.
Collapse
Affiliation(s)
- Ji-Fei Liu
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Li-Xia Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Juan-Ping Zhou
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Jia-Jia Zhang
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Qiong-Hui Wu
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Wei Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - De-Yi Chen
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China
| | - Zhen-Chang Zhang
- Department of Neurology, Lanzhou University Second Hospital, No.82, Cuiyingmen, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
59
|
Santo BA, Ciecierska SSK, Mousavi Janbeh Sarayi SM, Jenkins TD, Baig AA, Monteiro A, Koenigsknecht C, Pionessa D, Gutierrez L, King RM, Gounis M, Siddiqui AH, Tutino VM. Tectonic infarct analysis: A computational tool for automated whole-brain infarct analysis from TTC-stained tissue. Heliyon 2023; 9:e14837. [PMID: 37025889 PMCID: PMC10070917 DOI: 10.1016/j.heliyon.2023.e14837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/06/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Background Infarct volume measured from 2,3,5-triphenyltetrazolium chloride (TTC)-stained brain slices is critical to in vivo stroke models. In this study, we developed an interactive, tunable, software that automatically computes whole-brain infarct metrics from serial TTC-stained brain sections. Methods Three rat ischemic stroke cohorts were used in this study (Total n = 91 rats; Cohort 1 n = 21, Cohort 2 n = 40, Cohort 3 n = 30). For each, brains were serially-sliced, stained with TTC and scanned on both anterior and posterior sides. Ground truth annotation and infarct morphometric analysis (e.g., brain-Vbrain, infarct-Vinfarct, and non-infarct-Vnon-infarct volumes) were completed by domain experts. We used Cohort 1 for brain and infarct segmentation model development (n = 3 training cases with 36 slices [18 anterior and posterior faces], n = 18 testing cases with 218 slices [109 anterior and posterior faces]), as well as infarct morphometrics automation. The infarct quantification pipeline and pre-trained model were packaged as a standalone software and applied to Cohort 2, an internal validation dataset. Finally, software and model trainability were tested as a use-case with Cohort 3, a dataset from a separate institute. Results Both high segmentation and statistically significant quantification performance (correlation between manual and software) were observed across all datasets. Segmentation performance: Cohort 1 brain accuracy = 0.95/f1-score = 0.90, infarct accuracy = 0.96/f1-score = 0.89; Cohort 2 brain accuracy = 0.97/f1-score = 0.90, infarct accuracy = 0.97/f1-score = 0.80; Cohort 3 brain accuracy = 0.96/f1-score = 0.92, infarct accuracy = 0.95/f1-score = 0.82. Infarct quantification (cohort average): Vbrain (ρ = 0.87, p < 0.001), Vinfarct (0.92, p < 0.001), Vnon-infarct (0.80, p < 0.001), %infarct (0.87, p = 0.001), and infarct:non-infact ratio (ρ = 0.92, p < 0.001). Conclusion Tectonic Infarct Analysis software offers a robust and adaptable approach for rapid TTC-based stroke assessment.
Collapse
|
60
|
Sorbie A, Delgado Jiménez R, Weiler M, Benakis C. Protocol for microbiota analysis of a murine stroke model. STAR Protoc 2023; 4:101969. [PMID: 36625216 PMCID: PMC9843484 DOI: 10.1016/j.xpro.2022.101969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/26/2022] [Accepted: 12/09/2022] [Indexed: 01/11/2023] Open
Abstract
Investigations on the microbiota in neurological diseases such as stroke are increasingly common; however, stroke researchers may have limited experience with designing such studies. Here, we describe a protocol to conduct a stroke microbiota study in mice, from experimental stroke surgery and sample collection to data analysis. We provide details on sample processing and sequencing and provide a reproducible data analysis pipeline. In doing so, we hope to enable researchers to conduct robust studies and facilitate identification of stroke-associated microbial signatures. For complete details on the use and execution of this protocol, please refer to Sorbie et al. (2022).1.
Collapse
Affiliation(s)
- Adam Sorbie
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Rosa Delgado Jiménez
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Monica Weiler
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Corinne Benakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
61
|
Mottahedin A, Prag HA, Dannhorn A, Mair R, Schmidt C, Yang M, Sorby-Adams A, Lee JJ, Burger N, Kulaveerasingam D, Huang MM, Pluchino S, Peruzzotti-Jametti L, Goodwin R, Frezza C, Murphy MP, Krieg T. Targeting succinate metabolism to decrease brain injury upon mechanical thrombectomy treatment of ischemic stroke. Redox Biol 2023; 59:102600. [PMID: 36630820 PMCID: PMC9841348 DOI: 10.1016/j.redox.2023.102600] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/15/2022] [Accepted: 01/02/2023] [Indexed: 01/04/2023] Open
Abstract
Current treatments for acute ischemic stroke aim to reinstate a normal perfusion in the ischemic territory but can also cause significant ischemia-reperfusion (IR) injury. Previous data in experimental models of stroke show that ischemia leads to the accumulation of succinate, and, upon reperfusion, the accumulated succinate is rapidly oxidized by succinate dehydrogenase (SDH) to drive superoxide production at mitochondrial complex I. Despite this process initiating IR injury and causing further tissue damage, the potential of targeting succinate metabolism to minimize IR injury remains unexplored. Using both quantitative and untargeted high-resolution metabolomics, we show a time-dependent accumulation of succinate in both human and mouse brain exposed to ischemia ex vivo. In a mouse model of ischemic stroke/mechanical thrombectomy mass spectrometry imaging (MSI) shows that succinate accumulation is confined to the ischemic region, and that the accumulated succinate is rapidly oxidized upon reperfusion. Targeting succinate oxidation by systemic infusion of the SDH inhibitor malonate upon reperfusion leads to a dose-dependent decrease in acute brain injury. Together these findings support targeting succinate metabolism upon reperfusion to decrease IR injury as a valuable adjunct to mechanical thrombectomy in ischemic stroke.
Collapse
Affiliation(s)
- Amin Mottahedin
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK; Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.
| | - Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge University Hospitals, Cambridge, UK
| | - Andreas Dannhorn
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R & D, AstraZeneca, Cambridge, UK
| | - Richard Mair
- Division of Neurosurgery, Department of Clinical Neurosciences, Cambridge University Hospitals, Cambridge, UK
| | - Christina Schmidt
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Ming Yang
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Annabel Sorby-Adams
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Jordan J Lee
- Department of Medicine, University of Cambridge, Cambridge University Hospitals, Cambridge, UK
| | - Nils Burger
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Margaret M Huang
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, UK
| | - Richard Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R & D, AstraZeneca, Cambridge, UK
| | - Christian Frezza
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge University Hospitals, Cambridge, UK.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge University Hospitals, Cambridge, UK.
| |
Collapse
|
62
|
Automatic Cerebral Hemisphere Segmentation in Rat MRI with Ischemic Lesions via Attention-based Convolutional Neural Networks. Neuroinformatics 2023; 21:57-70. [PMID: 36178571 PMCID: PMC9931784 DOI: 10.1007/s12021-022-09607-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 10/14/2022]
Abstract
We present MedicDeepLabv3+, a convolutional neural network that is the first completely automatic method to segment cerebral hemispheres in magnetic resonance (MR) volumes of rats with ischemic lesions. MedicDeepLabv3+ improves the state-of-the-art DeepLabv3+ with an advanced decoder, incorporating spatial attention layers and additional skip connections that, as we show in our experiments, lead to more precise segmentations. MedicDeepLabv3+ requires no MR image preprocessing, such as bias-field correction or registration to a template, produces segmentations in less than a second, and its GPU memory requirements can be adjusted based on the available resources. We optimized MedicDeepLabv3+ and six other state-of-the-art convolutional neural networks (DeepLabv3+, UNet, HighRes3DNet, V-Net, VoxResNet, Demon) on a heterogeneous training set comprised by MR volumes from 11 cohorts acquired at different lesion stages. Then, we evaluated the trained models and two approaches specifically designed for rodent MRI skull stripping (RATS and RBET) on a large dataset of 655 MR rat brain volumes. In our experiments, MedicDeepLabv3+ outperformed the other methods, yielding an average Dice coefficient of 0.952 and 0.944 in the brain and contralateral hemisphere regions. Additionally, we show that despite limiting the GPU memory and the training data, our MedicDeepLabv3+ also provided satisfactory segmentations. In conclusion, our method, publicly available at https://github.com/jmlipman/MedicDeepLabv3Plus , yielded excellent results in multiple scenarios, demonstrating its capability to reduce human workload in rat neuroimaging studies.
Collapse
|
63
|
Ma R, Lu D, Xie Q, Yuan J, Ren M, Li Y, Wang J, Li J, Xu Z, Wang J. l-Borneol and d-Borneol promote transdifferentiation of astrocytes into neurons in rats by regulating Wnt/Notch pathway to exert neuroprotective effect during recovery from cerebral ischaemia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154583. [PMID: 36610167 DOI: 10.1016/j.phymed.2022.154583] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/18/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND The Chinese medicines Borneolum and l-Borneolum have neuroprotective effects on acute cerebral ischaemia-reperfusion (IR) in rats. Research on their effects during recovery from cerebral IR is lacking. Cerebral ischaemia can activate astrocytes for conversion into neurons. Neurogenesis cannot be achieved without nutritional support from an improved brain microenvironment through the blood circulation. PURPOSE The purpose of this study was to determine whether Borneolum and l-Borneolum can promote transdifferentiation of astrocytes into neurons by regulating the Wnt/Notch pathway to exert neuroprotective effects during recovery from cerebral ischaemia. STUDY DESIGN AND METHODS A suture crossing the external carotid artery to occlude the middle cerebral artery was used to prepare a model of cerebral IR (Longa et al., 1989). The Longa neurological function score, modified neurological severity score, tape removal test and grid misstep experiment were used to evaluate motor nerve function. Triphenyltetrazolium chloride was used to determine the extent of cerebral infarction. Left/right hemisphere contrast was used to measure brain atrophy. Astrocytes labelled with adeno-associated virus were used to track their fate after transdifferentiation. Laser speckle contrast imaging was used to observe the effects of l-Borneolum and Borneolum on cerebral blood flow. Immunofluorescence and western blotting were used to investigate their mechanisms. RESULTS l-Borneolum and Borneolum significantly improved neurological function and limb movement in rats with cerebral ischaemia during recovery and increased cerebral blood flow. l-Borneolum improved forelimb motor coordination more effectively than Borneolum and promoted transdifferentiation of astrocytes to GABAergic neurons in the striatal region. The expression of Wnt3a and Notch-1 was downregulated. The expression of vascular endothelial growth factor was not significantly changed. Borneolum improved forelimb sensitivity and alleviated cerebral infarction and brain atrophy more effectively than l-Borneolum, which promoted transdifferentiation of astrocytes into neurons and nestin expression and neurogenesis in the striatal zone. The expression of glycogen synthase kinase-3β and β-catenin was upregulated. l-Borneolum and Borneolum had no significant neuroprotective effect on the cortex and hippocampus. CONCLUSIONS l-Borneolum and Borneolum exerted neuroprotective effects on cerebral ischaemia during recovery by promoting neurogenesis and blood circulation in the striatal and subventricular zones. Their mechanisms may be related to the Wnt3a and Notch-1 pathways.
Collapse
Affiliation(s)
- Rong Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Medicine, Foshan University, Foshan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Danni Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Medicine, Foshan University, Foshan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianmei Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mihong Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiajun Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinxiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
64
|
Wong A, Bhuiyan MIH, Rothman J, Drew K, Pourrezaei K, Sun D, Barati Z. Near infrared spectroscopy detection of hemispheric cerebral ischemia following middle cerebral artery occlusion in rats. Neurochem Int 2023; 162:105460. [PMID: 36455748 PMCID: PMC10263189 DOI: 10.1016/j.neuint.2022.105460] [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: 08/31/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
Timely and sensitive in vivo estimation of ischemic stroke-induced brain infarction are necessary to guide diagnosis and evaluation of treatments' efficacy. The gold standard for estimation of the cerebral infarction volume is magnetic resonance imaging (MRI), which is expensive and not readily accessible. Measuring regional cerebral blood flow (rCBF) with Laser Doppler flowmetry (LDF) is the status quo for confirming reduced blood flow in experimental ischemic stroke models. However, rCBF reduction following cerebral artery occlusion often does not correlate with subsequent infarct volume. In the present study, we employed the continuous-wave near infrared spectroscopy (NIRS) technique to monitor cerebral oxygenation during 90 min of the intraluminal middle cerebral artery occlusion (MCAO) in Sprague-Dawley rats (n = 8, male). The NIRS device consisted of a controller module and an optical sensor with two LED light sources and two photodiodes making up two parallel channels for monitoring left and right cerebral hemispheres. Optical intensity measurements were converted to deoxyhemoglobin (Hb) and oxyhemoglobin (HbO2) changes relative to a 2-min window prior to MCAO. Area under the curve (auc) for Hb and HbO2 was calculated for the 90-min occlusion period for each hemisphere (ipsilateral and contralateral). To obtain a measure of total ischemia, auc of the contralateral side was subtracted from the ipsilateral side resulting in ΔHb and ΔHbO2 parameters. Infarct volume (IV) was calculated by triphenyl tetrazolium chloride (TTC) staining at 24h reperfusion. Results showed a significant negative correlation (r = -0.81, p = 0.03) between ΔHb and infarct volume. In conclusion, our results show feasibility of using a noninvasive optical imaging instrument, namely NIRS, in monitoring cerebral ischemia in a rodent stroke model. This cost-effective, non-invasive technique may improve the rigor of experimental models of ischemic stroke by enabling in vivo longitudinal assessment of cerebral oxygenation and ischemic injury.
Collapse
Affiliation(s)
- Ardy Wong
- Drexel University School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA, USA
| | - Mohammad Iqbal Hossain Bhuiyan
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, PA, 15260, USA; Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Education and Clinical Center, Pennsylvania, PA, 15260, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, 79968, USA
| | | | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, USA
| | - Kambiz Pourrezaei
- Drexel University School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, PA, 15260, USA; Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Education and Clinical Center, Pennsylvania, PA, 15260, USA
| | - Zeinab Barati
- Barati Medical LLC, Fairbanks, AK, USA; Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, USA.
| |
Collapse
|
65
|
Optimizing intraluminal monofilament model of ischemic stroke in middle-aged Sprague-Dawley rats. BMC Neurosci 2022; 23:75. [PMID: 36494808 PMCID: PMC9733327 DOI: 10.1186/s12868-022-00764-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Intraluminal monofilament model of middle cerebral artery occlusion (MCAO) is widely adopted for ischemic stroke; and Sprague-Dawley (SD) rats are commonly used rodents for preclinical research. Due to the paucity of information on the appropriate monofilament size for inducing MCAO in SD rats and the importance of including middle-aged models in ischemic stroke studies, we aimed to: (i). determine an appropriate Doccol® monofilament size for middle-aged male SD rats which weighed > 500 g following 24-h transient MCAO survival as well as (ii). demonstrate the optimal Doccol® filament size for middle-aged males (≤ 500 g) and females (273-300 g) while using young adult male SD rats (372-472 g) as control for severity of infarct volume following 7-days post-MCAO. All rats were subjected to 90-min transient MCAO. We show that 0.43 mm Doccol® monofilament size is more appropriate to induce large infarct lesion and optimal functional deficit when compared to 0.45 mm and 0.47 mm at 24 h post-MCAO. Our data on infarct volumes at 7 days post-MCAO as well as the observed weight loss and functional deficits at post-MCAO days 1, 3 and 7 demonstrate that 0.41 mm, 0.37 mm and 0.39 mm are optimal Doccol® filament sizes for middle-aged male (477.3 ± 39.61 g) and female (302.6 ± 26.28 g) as well as young-adult male (362.2 ± 28.38 g) SD rats, respectively.
Collapse
|
66
|
Khanaki K, Abedinzade M, Daei S. Effects of Urtica dioica extract on caspase-3 and cyclooxygenase-2 genes expression in a rat stroke model. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
67
|
Zhang L, Wang L, Hu P, Zhong X, Zhang D, Qiu F, Huang Z. Pinus massoniana needle extracts attenuate oxidative stress injury in cerebral ischemia reperfusion rats by regulating JNK3/caspase-3 signal transduction. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:563-572. [PMID: 36581582 PMCID: PMC10264975 DOI: 10.3724/zdxbyxb-2022-0326] [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: 06/20/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the effect and mechanism of Pinus massoniana needle extracts (PNE) on oxidative stress injury in cerebral ischemia reperfusion rats. METHODS The SD male rats were randomly divided into sham group, model control group, Edaravone (3 mg/kg) group, PNE low-dose (200 mg/kg), medium-dose (400 mg/kg) and high-dose (800 mg/kg) groups. PNE was administered by gavage for 7 d before modeling and 6 h after modeling in PNE treatment groups; Edaravone was given by intraperitoneal injection 7 d before modeling and 6 h after reperfusion. The rat model of cerebral ischemia reperfusion injury was established by middle cerebral artery occlusion method. After 24 h of reperfusion, the neurological deficit score, brain water content and cerebral infarction volume of rats were measured. The pathological changes of cerebral cortex and hippocampus were observed by HE staining, and the number of normal nerve cells was counted. The apoptosis rate of neurons in cerebral cortex was detected by TUNEL method. The content of nitric oxide (NO), malondialdehyde (MDA) and superoxide dismutase (SOD) activity in ischemic brain tissue were detected. The protein expression of c-Jun N-terminal kinase (JNK) 3, phosphorylated JNK3 (p-JNK3), B-cell lymphoma protein(Bcl) -2, Bcl-2 associated X (Bax), cytochrome C and caspase-3 in cerebral cortex were detected by Western blotting method. RESULTS Compared with the model control group, the behavioral score, brain water content and cerebral infarction volume in PNE groups were significantly reduced (all P<0.05), the pathological damage of cerebral cortex and hippocampal CA1 area was significantly alleviated, and the number of normal nerve cells in ischemic cortex and hippocampal CA1 area was increased (all P<0.05). The medium-dose PNE group had the best effect. Compared with the model control group, the apoptosis rate of cortical neurons, the content of NO and MDA in cerebral cortex, the ratio of p-JNK3/JNK3, the expression level of cytochrome C and caspase-3 protein in PNE medium-dose group were significantly reduced , and the activity of SOD, the Bcl-2/Bax ratio were significantly improved (all P<0.05). CONCLUSION PNE ameliorates brain injury after cerebral ischemia reperfusion in rats, which may be related to scavenging NO and MDA, inhibiting oxidative stress-mediated JNK3/caspase-3 signsal transduction to inhibit neuronal apoptosis.
Collapse
Affiliation(s)
- Lei Zhang
- 1. School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lingfeng Wang
- 1. School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Peihao Hu
- 1. School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaoming Zhong
- 1. School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Danni Zhang
- 1. School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Fengmei Qiu
- 1. School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhen Huang
- 1. School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- 2. Zhejiang Provincial Key Laboratory of Neuropharmacology and Translational Medicine, Hangzhou 310053, China
| |
Collapse
|
68
|
Zhao Y, Ding M, Yan F, Yin J, Shi W, Yang N, Zhao H, Fang Y, Huang Y, Zheng Y, Yang X, Li W, Ji X, Luo Y. Inhibition of the JAK2/STAT3 pathway and cell cycle re-entry contribute to the protective effect of remote ischemic pre-conditioning of rat hindlimbs on cerebral ischemia/reperfusion injury. CNS Neurosci Ther 2022; 29:866-877. [PMID: 36419252 PMCID: PMC9928551 DOI: 10.1111/cns.14023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022] Open
Abstract
AIMS Remote ischemic pre-conditioning (RIPC) protects against ischemia/reperfusion (I/R) injury. However, the mechanisms underlying this protection remain unclear. In the present study, we investigated the role of Janus-activated kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway and cell cycle arrest, and their relationship with neuronal apoptosis following RIPC. METHODS A rat cerebral I/R injury model was induced by middle cerebral artery occlusion (MCAO), and AG490 was used to investigate the mechanisms of RIPC. p-JAK2-, p-STAT3-, cyclin D1-, and cyclin-dependent kinase 6 (CDK6) expression was assessed by Western blotting and immunofluorescence staining. RESULTS RIPC reduced the infarct volume, improved neurological function, and increased neuronal survival. Furthermore, p-JAK2 and p-STAT3 were detected during the initial phase of reperfusion; the expression levels were significantly increased at 3 and 24 h after reperfusion and were suppressed by RIPC. Additionally, the MCAO-induced upregulation of the cell cycle regulators cyclin D1 and CDK6 was ameliorated by RIPC. Meanwhile, cyclin D1 and CDK6 were colocalized with p-STAT3 in the ischemic brain. CONCLUSION RIPC ameliorates the induction of the JAK2/STAT3 pathway and cell cycle regulators cyclin D1 and CDK6 by MCAO, and this net inhibition of cell cycle re-entry by RIPC is associated with downregulation of STAT3 phosphorylation.
Collapse
Affiliation(s)
- Yongmei Zhao
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Mao Ding
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Feng Yan
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Jie Yin
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Wenjuan Shi
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Nan Yang
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Haiping Zhao
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Yalan Fang
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Yuyou Huang
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Yangmin Zheng
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Xueqi Yang
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Wei Li
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina
| | - Xunming Ji
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina,Beijing Institute for Brain DisordersCapital Medical UniversityBeijingChina
| | - Yumin Luo
- Institute of Cerebrovascular Disease ResearchXuanwu Hospital of Capital Medical UniversityBeijingChina,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseasesBeijingChina,Beijing Institute for Brain DisordersCapital Medical UniversityBeijingChina
| |
Collapse
|
69
|
Koehn LM, Nguyen K, Chen X, Santoso A, Tucker R, Lim YP, Stonestreet BS. Effects of Three Different Doses of Inter-Alpha Inhibitor Proteins on Severe Hypoxia-Ischemia-Related Brain Injury in Neonatal Rats. Int J Mol Sci 2022; 23:13473. [PMID: 36362257 PMCID: PMC9655902 DOI: 10.3390/ijms232113473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2024] Open
Abstract
Hypoxia-ischemia (HI)-related brain injury is an important cause of morbidity and long-standing disability in newborns. We have previously shown that human plasma-derived inter-alpha inhibitor proteins (hIAIPs) attenuate HI-related brain injury in neonatal rats. The optimal dose of hIAIPs for their neuroprotective effects and improvement in behavioral outcomes remains to be determined. We examined the efficacy of 30, 60, or 90 mg/kg of hIAIPs administered to neonatal rats after exposure to HI for 2 h. Postnatal day 7 (P7) Wistar rats were exposed to either sham-surgery or unilateral HI (right carotid artery ligation, 2 h of 8% O2) brain injury. A placebo, 30, 60, or 90 mg/kg of hIAIPs were injected intraperitoneally at 0, 24 and 48 h after HI (n = 9-10/sex). We carried out the following behavioral analyses: P8 (righting reflex), P9 (negative geotaxis) and P10 (open-field task). Rats were humanely killed on P10 and their brains were stained with cresyl violet. Male extension/contraction responses and female righting reflex times were higher in the HI placebo groups than the sham groups. Female open-field exploration was lower in the HI placebo group than the sham group. hIAIPs attenuated these behavioral deficits. However, the magnitude of the responses did not vary by hIAIP dose. hIAIPs reduced male brain infarct volumes in a manner that correlated with improved behavioral outcomes. Increasing the hIAIP dose from 30 to 90 mg/kg did not further accentuate the hIAIP-related decreases in infarct volumes. We conclude that larger doses of hIAIPs did not provide additional benefits over the 30 mg/kg dose for behavior tasks or reductions in infarct volumes in neonatal rats after exposure to severe HI.
Collapse
Affiliation(s)
- Liam M. Koehn
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Kevin Nguyen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Xiaodi Chen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | | | - Richard Tucker
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02905, USA
| | - Barbara S. Stonestreet
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
70
|
Toti KS, Verma R, McGonnigle MJ, Gamiotea Turro D, Wen Z, Lewicki SA, Liang BT, Jacobson KA. Structure-Activity Relationship and Neuroprotective Activity of 1,5-Dihydro-2 H-naphtho[1,2- b][1,4]diazepine-2,4(3 H)-diones as P2X4 Receptor Antagonists. J Med Chem 2022; 65:13967-13987. [PMID: 36150180 PMCID: PMC9653265 DOI: 10.1021/acs.jmedchem.2c01197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We analyzed the P2X4 receptor structure-activity relationship of a known antagonist 5, a 1,5-dihydro-2H-naphtho[1,2-b][1,4]diazepine-2,4(3H)-dione. Following extensive modification of the reported synthetic route, 4-pyridyl 21u (MRS4719) and 6-methyl 22c (MRS4596) analogues were most potent at human (h) P2X4R (IC50 0.503 and 1.38 μM, respectively, and selective versus hP2X1R, hP2X2/3R, hP2X3R). Thus, the naphthalene 6-, but not 7-position was amenable to substitution, and an N-phenyl ring aza-scan identified 21u with 3-fold higher activity than 5. Compounds 21u and 22c showed neuroprotective and learning- and memory-enhancing activities in a mouse middle cerebral artery occlusion (MCAO) model of ischemic stroke, with potency of 21u > 22c. 21u dose-dependently reduced infarct volume and reduced brain atrophy at 3 and 35 days post-stroke, respectively. Relevant to clinical implication, 21u also reduced ATP-induced [Ca2+]i influx in primary human monocyte-derived macrophages. This study indicates the translational potential of P2X4R antagonists for treating ischemic stroke, including in aging populations.
Collapse
Affiliation(s)
- Kiran S Toti
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland 20892-0810, United States
| | - Rajkumar Verma
- Department of Neuroscience, UConn School of Medicine, Farmington, Connecticut 06032, United States
| | - Michael J McGonnigle
- Department of Neuroscience, UConn School of Medicine, Farmington, Connecticut 06032, United States
| | - Daylin Gamiotea Turro
- Department of Neuroscience, UConn School of Medicine, Farmington, Connecticut 06032, United States
| | - Zhiwei Wen
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland 20892-0810, United States
| | - Sarah A Lewicki
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland 20892-0810, United States
| | - Bruce T Liang
- Calhoun Cardiology Center, UConn School of Medicine, Farmington, Connecticut 06032, United States
| | - Kenneth A Jacobson
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland 20892-0810, United States
| |
Collapse
|
71
|
Lu H, Ashiqueali R, Lin CI, Walchale A, Clendaniel V, Matheson R, Fisher M, Lo EH, Selim M, Shehadah A. Histone Deacetylase 3 Inhibition Decreases Cerebral Edema and Protects the Blood–Brain Barrier After Stroke. Mol Neurobiol 2022; 60:235-246. [DOI: 10.1007/s12035-022-03083-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022]
Abstract
AbstractWe have previously shown that selective inhibition of histone deacetylase 3 (HDAC3) decreases infarct volume and improves long-term functional outcomes after stroke. In this study, we examined the effects of HDAC3 inhibition on cerebral edema and blood–brain barrier (BBB) leakage and explored its underlying mechanisms. Adult male Wistar rats were subjected to 2-h middle cerebral artery occlusion (MCAO) and randomly treated i.p. with either vehicle or a selective HDAC3 inhibitor (RGFP966) at 2 and 24 h after stroke. Modified neurological severity scores (mNSS) were calculated at 2 h, 1 day, and 3 days. H&E, Evans blue dye (EBD) assay, and fluorescein isothiocyanate (FITC)-dextran were employed to assess cerebral edema and BBB leakage. Western blot for matrix metalloproteinase-9 (MMP9), MMP-9 zymography, and immunostaining for HDAC3, GFAP, Iba-1, albumin, aquaporin-4, claudin-5, ZO-1, and NF-kB were performed. Early RGFP966 administration decreased cerebral edema (p = 0.002) and BBB leakage, as measured by EBD assay, FITC-dextran, and albumin extravasation (p < 0.01). RGFP966 significantly increased tight junction proteins (claudin-5 and ZO-1) in the peri-infarct area. RGFP966 also significantly decreased HDAC3 in GFAP + astrocytes, which correlated with better mNSS (r = 0.67, p = 0.03) and decreased cerebral edema (r = 0.64, p = 0.04). RGFP966 decreased aquaporin-4 in GFAP + astrocytes (p = 0.002), as well as, the inflammatory markers Iba-1, NF-kB, and MMP9 in the ischemic brain (p < 0.05). Early HDAC3 inhibition decreases cerebral edema and BBB leakage. BBB protection by RGFP966 is mediated in part by the upregulation of tight junction proteins, downregulation of aquaporin-4 and HDAC3 in astrocytes, and decreased neuroinflammation.
Collapse
|
72
|
Chen G, Yan S, Zhang J, Zhang J, Deng IB, He R. The alternative 3' splice site of GPNMB may promote neuronal survival after neonatal hypoxic-ischemic encephalopathy injury. IBRAIN 2022; 8:302-313. [PMID: 37786733 PMCID: PMC10529014 DOI: 10.1002/ibra.12056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 10/04/2023]
Abstract
This study aimed to decipher the effect of glycoprotein nonmetastatic melanoma protein B (GPNMB) on neonatal hypoxic-ischemic encephalopathy (NHIE) and its potential molecular mechanism. The hypoxic-ischemic (HI) model was established in 7-day-old rats, and then, Zea-Longa scores and Nissl staining were performed to measure brain damage post-HI. In addition, gene sequencing was used to detect the differential expression genes (DEGs), and then, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases were used to determine the function of DEGs. Furthermore, an oxygen-glucose deprivation (OGD) model was developed in SY5Y cells and human fetal neurons, and then, the level of GPNMB was verified by quantitative real-time polymerase chain reaction. In addition, methyl thiazolyl tetrazolium and cell counting kit-8 assays were applied after GPNMB interference. Finally, the alternative splicing of GPNMB expression was analyzed using Splice Grapher software. The results indicated that HI induced marked neurological impairment and neuron injury in rats. Also, GPNMB was the most obviously upregulated gene in DEGs. Additionally, GPNMB was upregulated significantly in SY5Y and fetal neurons after OGD, and GPNMB-si promoted an increase in cell viability and number. Moreover, we found that the GPNMB alternative splicing type was the Alternative 3' splice site, with the alternative splicing site in 143382985:143404102. Herein, GPNMB promotes a crucial regulatory mechanism with alternative splicing for neuronal survival after NHIE.
Collapse
Affiliation(s)
- Guo‐Jiao Chen
- Bioinformatics CenterKunming Medical UniversityKunmingYunnanChina
| | | | | | - Ji Zhang
- Southwest Medical UniversityLuzhouSichuanChina
| | - Isaac Bul Deng
- Center for Epogenetics and Induced Pluripotent Stem Cells, Kennedy Krieger InstituteJohns Hopkins UniversityBaltimoreUSA
| | - Rong He
- Bioinformatics CenterKunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
73
|
Guo Y, Zuo W, Yin L, Gu T, Wang S, Fang Z, Wang B, Dong H, Hou W, Zuo Z, Deng J. Pioglitazone Attenuates Ischemic Stroke Aggravation By Blocking PPARγ Reduction and Inhibiting Chronic Inflammation in Diabetic Mice. Eur J Neurosci 2022; 56:4948-4961. [PMID: 35945686 DOI: 10.1111/ejn.15789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/03/2022] [Indexed: 11/28/2022]
Abstract
Diabetes can cause vascular remodeling and is associated with worse outcome after ischemic stroke. Pioglitazone is a commonly used anti-diabetic agent. However, it is not known whether pioglitazone use before ischemia could reduce brain ischemic injury. Pioglitazone was administered to 5-week-old db+ or db/db mice. Cerebral vascular remodeling was examined at the age of 9 weeks. Expression of peroxisome proliferator-activated receptor-γ (PPARγ), p-PPARγ (S112 and S273), nucleotide-binding domain (NOD)-like receptor protein 3 (Nlrp3), interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) was evaluated in the somatosensory cortex of mice. Neurological outcome was evaluated 24 h after brain ischemia. Results showed that early pioglitazone treatment provided a long-lasting effect of euglycemia but enhanced hyperlipidemia in the db/db mice. Diabetic mice exhibited increased vascular tortuosity, narrower middle cerebral artery (MCA) width and IgG leakage in the brain. These changes were blocked by early pioglitazone treatment. In diabetic animals, PPARγ expression was reduced and p-PPARγ at S273 but not S112, Nlrp3, IL-1β and TNF-α were increased in the somatosensory cortex. PPARγ decrease and Nlrp3 increase were mainly in the neurons of the diabetic brain, which was reversed by early pioglitazone treatment. Pioglitazone attenuated the aggravated neurological outcome after stroke in diabetic mice. But this protective effect was abolished through restoring cerebral inflammation by intracerebroventricular administration of IL-1β and TNF-α in pioglitazone treated diabetic mice before MCAO. In summary, early pioglitazone treatment attenuates cerebral vascular remodeling and ischemic brain injury possibly via blocking chronic neuroinflammation in the db/db mice.
Collapse
Affiliation(s)
- Yaru Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenqiang Zuo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lu Yin
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tingting Gu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zongping Fang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bairen Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States
| | - Jiao Deng
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
74
|
Schacke S, Kirkpatrick J, Stocksdale A, Bauer R, Hagel C, Riecken LB, Morrison H. Ezrin deficiency triggers glial fibrillary acidic protein upregulation and a distinct reactive astrocyte phenotype. Glia 2022; 70:2309-2329. [PMID: 35929192 DOI: 10.1002/glia.24253] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 01/02/2023]
Abstract
Astrocytes are increasingly being recognized as contributors to physiological brain function and behavior. Astrocytes engage in glia-synaptic interactions through peripheral astrocyte processes, thus modulating synaptic signaling, for example, by handling glutamate removal from the synaptic cleft and (re)provision to axonal terminals. Peripheral astrocyte processes are ultrafine membrane protrusions rich in the membrane-to-actin cytoskeleton linker Ezrin, an essential component of in vitro filopodia formation and in vivo peripheral astrocyte process motility. Consequently, it has been postulated that Ezrin significantly contributes to neurodevelopment as well as astrocyte functions within the adult brain. However, while Ezrin has been studied in vitro within cultured primary astrocytes, in vivo studies on the role of Ezrin in astrocytes remain to be conducted and consequences of its depletion to be studied. Here, we investigated consequences of Ezrin deletion in the mouse brain starting from early neuronal specification. While Ezrin knockout did not impact prenatal cerebral cortex development, behavioral phenotyping depicted reduced exploratory behavior. Starting with postnatal appearance of glia cells, Ezrin was verified to remain predominantly expressed in astrocytes. Proteome analysis of Ezrin deficient astrocytes revealed alterations in glutamate and ion homeostasis, metabolism and cell morphology - important processes for synaptic signal transmission. Notably, Ezrin deletion in astrocytes provoked (GFAP) glial fibrillary acidic protein upregulation - a marker of astrocyte activation and reactive astrogliosis. However, this spontaneous, reactive astrogliosis exhibited proteome changes distinct from ischemic-induced reactive astrogliosis. Moreover, in experimental ischemic stroke, Ezrin knockout mice displayed reduced infarct volume, indicating a protective effect of the Ezrin deletion-induced changes and astrogliosis.
Collapse
Affiliation(s)
- Stephan Schacke
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | | | - Amy Stocksdale
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Christian Hagel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Helen Morrison
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany.,Faculty of Biological Sciences, Friedrich-Schiller University, Jena, Germany
| |
Collapse
|
75
|
LU XY, ZHENG H, LÜ Q, LI Q, ZHANG H, TIAN H. Effects of acupuncture on Beclin-1 and Caspase-3 expression in hippocampal tissue of rats with cerebral ischemia-reperfusion injury: observations at different time points. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2022. [DOI: 10.1016/j.wjam.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
76
|
He J, Xue K, Liu J, Gu JH, Peng B, Xu L, Wang G, Jiang Z, Li X, Zhang Y. Timely and Appropriate Administration of Inhaled Argon Provides Better Outcomes for tMCAO Mice: A Controlled, Randomized, and Double-Blind Animal Study. Neurocrit Care 2022; 37:91-101. [PMID: 35137354 DOI: 10.1007/s12028-022-01448-9] [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: 10/07/2021] [Accepted: 01/10/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Inhaled argon (iAr) has shown promising therapeutic efficacy for acute ischemic stroke and has exhibited impressive advantages over other inert gases as a neuroprotective agent. However, the optimal dose, duration, and time point of iAr for acute ischemic stroke are unknown. Here, we explored variable iAr schedules and evaluated the neuroprotective effects of acute iAr administration on lesion volume, brain edema, and neurological function in a mouse model of cerebral ischemic/reperfusion injury. METHODS Adult ICR (Institute of Cancer Research) mice were randomly subjected to sham, moderate (1.5 h), or severe (3 h) transient middle cerebral artery occlusion (tMCAO). One hour after tMCAO, the mice were randomized to variable iAr protocols or air. General and focal deficit scores were assessed during double-blind treatment. Infarct volume, overall recovery, and brain edema were analyzed 24 h after cerebral ischemic/reperfusion injury. RESULTS Compared with those in the tMCAO-only group, lesion volume (p < 0.0001) and neurologic outcome (general, p < 0.0001; focal, p < 0.0001) were significantly improved in the group administered iAr 1 h after stroke onset (during ischemia). Short-term argon treatment (1 or 3 h) significantly improved the infarct volume (1 vs. 24 h, p < 0.0001; 3 vs. 24 h, p < 0.0001) compared with argon inhalation for 24 h. The concentration of iAr was confirmed to be a key factor in improving focal neurological outcomes relative to that in the tMCAO group, with higher concentrations of iAr showing better effects. Additionally, even though ischemia research has shown an increase in cerebral damage proportional to the ischemia time, argon administration showed significant neuroprotective effects on infarct volume (p < 0.0001), neurological deficits (general, p < 0.0001; focal, p < 0.0001), weight recovery (p < 0.0001), and edema (p < 0.0001) in general, particularly in moderate stroke. CONCLUSIONS Timely iAr administration during ischemia showed optimal neurological outcomes and minimal infarct volumes. Moreover, an appropriate duration of argon administration was important for better neuroprotective efficacy. These findings may provide vital guidance for using argon as a neuroprotective agent and moving to clinical trials in acute ischemic stroke.
Collapse
Affiliation(s)
- Juan He
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Ke Xue
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jiayi Liu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jin-Hua Gu
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China
| | - Bin Peng
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Lihua Xu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Guohua Wang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Zhenglin Jiang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Xia Li
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China.
| | - Yunfeng Zhang
- Stroke Center and Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226019, Jiangsu, China.
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
77
|
Zhang K, Yang Y, Ge H, Wang J, Lei X, Chen X, Wan F, Feng H, Tan L. Neurogenesis and Proliferation of Neural Stem/Progenitor Cells Conferred by Artesunate via FOXO3a/p27Kip1 Axis in Mouse Stroke Model. Mol Neurobiol 2022; 59:4718-4729. [PMID: 35596896 DOI: 10.1007/s12035-021-02710-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/21/2021] [Indexed: 11/24/2022]
Abstract
Promoting neurogenesis and proliferation of endogenous neural stem/progenitor cells (NSPCs) is considered a promising strategy for neurorehabilitation after stroke. Our previous study revealed that a moderate dose of artesunate (ART, 150 mg/kg) could enhance functional recovery in middle cerebral artery occlusion (MCAO) mice. This study aimed to investigate the effects of ART treatment on neurogenesis and proliferation of NSPCs using a rodent MCAO model. MRI results indicated that the ischemic brain volume of MCAO mice was reduced by ART treatment. The results of diffusion tensor imaging, electron microscopic, and immunofluorescence of Tuj-1 also revealed that ischemia-induced white matter lesion was alleviated by ART treatment. After ischemia/reperfusion, the proportion of Brdu + endogenous NSPCs in the ipsilateral subventricular zone and peri-infarct cortex was increased by ART treatment. Furthermore, the neuro-restorative effects of ART were abolished by the overexpression of FOXO3a. These findings suggested that ART could rescue ischemia/reperfusion damage and alleviate white matter injury, subsequently contributing to post-stroke functional recovery by promoting neurogenesis and proliferation of endogenous NSPCs via the FOXO3a/p27Kip1 pathway.
Collapse
Affiliation(s)
- Kaiyuan Zhang
- Department of Neurosurgery, Southwest Hospital, the Third Military Medical University (Army Military Medical University), Chongqing, China
- Department of Neurosurgery, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, China
| | - Yang Yang
- Department of Neurosurgery, Southwest Hospital, the Third Military Medical University (Army Military Medical University), Chongqing, China
- Department of Neurosurgery, 904Th Hospital of the PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu, China
| | - Hongfei Ge
- Department of Neurosurgery, Southwest Hospital, the Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Ju Wang
- Department of Neurosurgery, Southwest Hospital, the Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xuejiao Lei
- Department of Neurosurgery, Southwest Hospital, the Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xuezhu Chen
- Department of Neurosurgery, Southwest Hospital, the Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Feng Wan
- Department of Electrical and Computer Engineering, Faculty of Science and Technology, University of Macau, Macau, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, the Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, the Third Military Medical University (Army Military Medical University), Chongqing, China.
- Department of Electrical and Computer Engineering, Faculty of Science and Technology, University of Macau, Macau, China.
| |
Collapse
|
78
|
Hsu PC, Lan YJ, Chen CC, Lee LY, Chen WP, Wang YC, Lee YH. Erinacine A attenuates glutamate transporter 1 downregulation and protects against ischemic brain injury. Life Sci 2022; 306:120833. [PMID: 35882273 DOI: 10.1016/j.lfs.2022.120833] [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: 03/09/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 11/27/2022]
Abstract
Maintaining glutamate homeostasis through astrocyte-enriched glutamate transporter 1 (GLT-1) is critical for neuronal survival, but it is often disrupted after brain injury. Hericium erinaceus (HE), an edible mushroom, was reported to be anti-inflammatory and neuroprotective against brain ischemia, but its effect on glutamate homeostasis was unknown. Here we investigated the neuroprotective effect of erinacine A (EA), an active component of HE, with special focus on the GLT-1 function in the in vitro and in vivo cerebral ischemia mouse models. By using oxygen-glucose deprivation (OGD) to challenge mouse glia-neuron (GN) mixed culture as the in vitro model, we found that EA treatment significantly improved neuronal/astroglial survival and attenuated OGD-induced proinflammatory NFκB and AKT signaling activations. Notably, EA attenuated OGD-induced GLT-1 downregulation, and a selective GLT-1 inhibitor WAY-213613 reversed these EA-mediated neuroprotection. EA also ameliorated glutamate excitotoxicity effectively. In a transient hypoxia-ischemia (tHI) brain injury mouse model, we examined an EA treatment strategy by performing a pre-tHI daily oral gavage of EA (oEA) for 7 days followed by a post-tHI intranasal injection of EA (nEA) for 3 days, and found that this treatment significantly protected sensorimotor cortex and improved the post-tHI forepaw grip strength. Western blotting results further revealed that EA treatment also preserved astrocyte-enriched glutamate and aspartate transporter (GLAST) as well as a GLT-1 function-associated potassium channel Kir4.1 in the cerebral cortex and striatum after tHI. These results suggest that EA is effective for preserving GLT-1 and glutamate clearance machinery to protect against excitotoxicity after ischemic brain injury.
Collapse
Affiliation(s)
- Pei-Chien Hsu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 112, Taiwan
| | - Yi-Jie Lan
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 112, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 324, Taiwan
| | - Li-Ya Lee
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 324, Taiwan
| | - Wan-Ping Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 324, Taiwan
| | - Yun-Ching Wang
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 112, Taiwan
| | - Yi-Hsuan Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 112, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei 112, Taiwan.
| |
Collapse
|
79
|
Karizmeh MS, Shabani M, Shabani M, Sardari M, Babaei JF, Nabavizadeh F, Sadr SS, Adeli S. Preconditioning exercise reduces hippocampal neuronal damage via increasing Klotho expression in ischemic rats. Brain Res Bull 2022; 188:133-142. [DOI: 10.1016/j.brainresbull.2022.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 01/24/2023]
|
80
|
Lopez MS, Morris-Blanco KC, Ly N, Maves C, Dempsey RJ, Vemuganti R. MicroRNA miR-21 Decreases Post-stroke Brain Damage in Rodents. Transl Stroke Res 2022; 13:483-493. [PMID: 34796453 PMCID: PMC11846127 DOI: 10.1007/s12975-021-00952-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022]
Abstract
Due to their role in controlling translation, microRNAs emerged as novel therapeutic targets to modulate post-stroke outcomes. We previously reported that miR-21 is the most abundantly induced microRNA in the brain of rodents subjected to preconditioning-induced cerebral ischemic tolerance. We currently show that intracerebral administration of miR-21 mimic decreased the infarct volume and promoted better motor function recovery in adult male and female C57BL/6 mice subjected to transient middle cerebral artery occlusion. The miR-21 mimic treatment is also efficacious in aged mice of both sexes subjected to focal ischemia. Mechanistically, miR-21 mimic treatment decreased the post-ischemic levels of several pro-apoptotic and pro-inflammatory RNAs, which might be responsible for the observed neuroprotection. We further observed post-ischemic neuroprotection in adult mice administered with miR-21 mimic intravenously. Overall, the results of this study implicate miR-21 as a promising candidate for therapeutic translation after stroke.
Collapse
Affiliation(s)
- Mary S Lopez
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
- Cell & Molecular Pathology Training Program, University of Wisconsin, Madison, WI, 53792, USA
| | | | - Nancy Ly
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Carly Maves
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Robert J Dempsey
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.
- Cell & Molecular Pathology Training Program, University of Wisconsin, Madison, WI, 53792, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
81
|
Yshii L, Pasciuto E, Bielefeld P, Mascali L, Lemaitre P, Marino M, Dooley J, Kouser L, Verschoren S, Lagou V, Kemps H, Gervois P, de Boer A, Burton OT, Wahis J, Verhaert J, Tareen SHK, Roca CP, Singh K, Whyte CE, Kerstens A, Callaerts-Vegh Z, Poovathingal S, Prezzemolo T, Wierda K, Dashwood A, Xie J, Van Wonterghem E, Creemers E, Aloulou M, Gsell W, Abiega O, Munck S, Vandenbroucke RE, Bronckaers A, Lemmens R, De Strooper B, Van Den Bosch L, Himmelreich U, Fitzsimons CP, Holt MG, Liston A. Astrocyte-targeted gene delivery of interleukin 2 specifically increases brain-resident regulatory T cell numbers and protects against pathological neuroinflammation. Nat Immunol 2022; 23:878-891. [PMID: 35618831 PMCID: PMC9174055 DOI: 10.1038/s41590-022-01208-z] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/11/2022] [Indexed: 12/21/2022]
Abstract
The ability of immune-modulating biologics to prevent and reverse pathology has transformed recent clinical practice. Full utility in the neuroinflammation space, however, requires identification of both effective targets for local immune modulation and a delivery system capable of crossing the blood-brain barrier. The recent identification and characterization of a small population of regulatory T (Treg) cells resident in the brain presents one such potential therapeutic target. Here, we identified brain interleukin 2 (IL-2) levels as a limiting factor for brain-resident Treg cells. We developed a gene-delivery approach for astrocytes, with a small-molecule on-switch to allow temporal control, and enhanced production in reactive astrocytes to spatially direct delivery to inflammatory sites. Mice with brain-specific IL-2 delivery were protected in traumatic brain injury, stroke and multiple sclerosis models, without impacting the peripheral immune system. These results validate brain-specific IL-2 gene delivery as effective protection against neuroinflammation, and provide a versatile platform for delivery of diverse biologics to neuroinflammatory patients.
Collapse
Affiliation(s)
- Lidia Yshii
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
| | - Emanuela Pasciuto
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
| | - Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Loriana Mascali
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Pierre Lemaitre
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Marika Marino
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
| | - James Dooley
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Lubna Kouser
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Stijn Verschoren
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
| | - Vasiliki Lagou
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Hannelore Kemps
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Pascal Gervois
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Antina de Boer
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
| | - Oliver T Burton
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Jérôme Wahis
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
| | - Jens Verhaert
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
| | - Samar H K Tareen
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Carlos P Roca
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Kailash Singh
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Carly E Whyte
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Axelle Kerstens
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
- VIB Bio-Imaging Core, Leuven, Belgium
| | | | | | - Teresa Prezzemolo
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Electrophysiology Expertise Unit, Leuven, Belgium
| | - Amy Dashwood
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Junhua Xie
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Eline Creemers
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Electrophysiology Expertise Unit, Leuven, Belgium
| | - Meryem Aloulou
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
- Toulouse Institute for Infectious and Inflammatory diseases (INFINITY), INSERM UMR1291, CNRS UMR 5051, Toulouse, France
| | - Willy Gsell
- KU Leuven, Department of Imaging and Pathology, Biomedical MRI, Leuven, Belgium
| | - Oihane Abiega
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Sebastian Munck
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
- VIB Bio-Imaging Core, Leuven, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Annelies Bronckaers
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Robin Lemmens
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
- University Hospitals Leuven, Department of Neurology, Leuven, Belgium
| | - Bart De Strooper
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
- Dementia Research Institute, University College London, London, United Kingdom
| | - Ludo Van Den Bosch
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven - Department of Neurosciences, Leuven, Belgium
| | - Uwe Himmelreich
- KU Leuven, Department of Imaging and Pathology, Biomedical MRI, Leuven, Belgium
| | - Carlos P Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Matthew G Holt
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven - Department of Neurosciences, Leuven, Belgium.
- Instituto de Investigaçāo e Inovaçāo em Saúde (i3S), University of Porto, Porto, Portugal.
| | - Adrian Liston
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium.
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom.
| |
Collapse
|
82
|
Rayasam A, Kijak JA, Kissel L, Choi YH, Kim T, Hsu M, Joshi D, Laaker CJ, Cismaru P, Lindstedt A, Kovacs K, Vemuganti R, Chiu SY, Priyathilaka TT, Sandor M, Fabry Z. CXCL13 expressed on inflamed cerebral blood vessels recruit IL-21 producing T FH cells to damage neurons following stroke. J Neuroinflammation 2022; 19:125. [PMID: 35624463 PMCID: PMC9145182 DOI: 10.1186/s12974-022-02490-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/12/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Ischemic stroke is a leading cause of mortality worldwide, largely due to the inflammatory response to brain ischemia during post-stroke reperfusion. Despite ongoing intensive research, there have not been any clinically approved drugs targeting the inflammatory component to stroke. Preclinical studies have identified T cells as pro-inflammatory mediators of ischemic brain damage, yet mechanisms that regulate the infiltration and phenotype of these cells are lacking. Further understanding of how T cells migrate to the ischemic brain and facilitate neuronal death during brain ischemia can reveal novel targets for post-stroke intervention. METHODS To identify the population of T cells that produce IL-21 and contribute to stroke, we performed transient middle cerebral artery occlusion (tMCAO) in mice and performed flow cytometry on brain tissue. We also utilized immunohistochemistry in both mouse and human brain sections to identify cell types and inflammatory mediators related to stroke-induced IL-21 signaling. To mechanistically demonstrate our findings, we employed pharmacological inhibitor anti-CXCL13 and performed histological analyses to evaluate its effects on brain infarct damage. Finally, to evaluate cellular mechanisms of stroke, we exposed mouse primary neurons to oxygen glucose deprivation (OGD) conditions with or without IL-21 and measured cell viability, caspase activity and JAK/STAT signaling. RESULTS Flow cytometry on brains from mice following tMCAO identified a novel population of cells IL-21 producing CXCR5+ CD4+ ICOS-1+ T follicular helper cells (TFH) in the ischemic brain early after injury. We observed augmented expression of CXCL13 on inflamed brain vascular cells and demonstrated that inhibition of CXCL13 protects mice from tMCAO by restricting the migration and influence of IL-21 producing TFH cells in the ischemic brain. We also illustrate that neurons express IL-21R in the peri-infarct regions of both mice and human stroke tissue in vivo. Lastly, we found that IL-21 acts on mouse primary ischemic neurons to activate the JAK/STAT pathway and induce caspase 3/7-mediated apoptosis in vitro. CONCLUSION These findings identify a novel mechanism for how pro-inflammatory T cells are recruited to the ischemic brain to propagate stroke damage and provide a potential new therapeutic target for stroke.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA.
| | - Julie A Kijak
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Lee Kissel
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Yun Hwa Choi
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Taehee Kim
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Martin Hsu
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Dinesh Joshi
- Department of Physiology, University of Wisconsin School of Medicine, Madison, WI, USA
| | - Collin J Laaker
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Peter Cismaru
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Anders Lindstedt
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Krisztian Kovacs
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Raghu Vemuganti
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Administration Hospital, Madison, WI, USA
| | - Shing Yan Chiu
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Physiology, University of Wisconsin School of Medicine, Madison, WI, USA
| | - Thanthrige Thiunuwan Priyathilaka
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
83
|
Kim S, Lee W, Jo H, Sonn SK, Jeong SJ, Seo S, Suh J, Jin J, Kweon HY, Kim TK, Moon SH, Jeon S, Kim JW, Kim YR, Lee EW, Shin HK, Park SH, Oh GT. The antioxidant enzyme Peroxiredoxin-1 controls stroke-associated microglia against acute ischemic stroke. Redox Biol 2022; 54:102347. [PMID: 35688114 PMCID: PMC9184746 DOI: 10.1016/j.redox.2022.102347] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 01/04/2023] Open
Affiliation(s)
- Sinai Kim
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Wonhyo Lee
- Department of Biological Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, South Korea
| | - Huiju Jo
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seong-Keun Sonn
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Se-Jin Jeong
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Seungwoon Seo
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Joowon Suh
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jing Jin
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Hyae Yon Kweon
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Tae Kyeong Kim
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Shin Hye Moon
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sejin Jeon
- Department of Biological Sciences and Biotechnology Major in Bio-Vaccine Engineering Andong National University, Andong, South Korea
| | - Jong Woo Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, South Korea; Department of Functional Genomics, University of Science and Technology (UST), Daejeon, South Korea
| | - Yu Ri Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine 1672, Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, South Korea; Department of Functional Genomics, University of Science and Technology (UST), Daejeon, South Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Sung Ho Park
- Department of Biological Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, South Korea.
| | - Goo Taeg Oh
- Heart-Immune-Brain Network Research Center, Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
84
|
Khanaki K, Fekri A, Abedinzade M, Mohammadi E, Aghajanpour F. Potential anti-inflammatory effect of Lamium album extract through caspase-3 and cyclooxygenase-2 genes expression in a rat model of middle cerebral artery occlusion. Folia Med (Plovdiv) 2022; 64:275-282. [DOI: 10.3897/folmed.64.e60562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/18/2021] [Indexed: 11/12/2022] Open
Abstract
Introduction: Stroke is one of the most common causes of death worldwide. Inflammation and apoptosis play an important role in the cascade of ischemic stroke.
Aim: The aim of the present study was to evaluate the pretreatment effects of Lamium album (L. album) extract on caspase-3 and cyclooxygenase-2 (COX-2) expression, infarct volume, and neurological deficit score in a rat model of middle cerebral artery occlusion (MCAO).
Materials and methods: Wistar male rats were randomly divided into three groups: 1) MCAO group (1 h after MCAO, reperfusion was allowed for 24 h by retracting the thread); 2) L. album + MCAO group [receiving L. album extract (100 mg/kg via intraperitoneal) for a week before MCAO]; 3) sham group. The expression level of caspase-3 and COX-2 in the core, penumbra, and subcortex regions was measured by real time-PCR technique. Infarct volume and neurological deficit score were also assessed.
Results: The mRNA expression of caspase-3 in the core, penumbra, and subcortex regions in L. album group was significantly reduced compared to MCAO group (p<0.05). Expression level of COX-2 in the subcortex of the rats exposed to L. album was statistically decreased relative to MCAO group (p<0.05). Infarct volume in the core, penumbra, and subcortex was significantly reduced in the L. album group compared with MCAO group (p<0.001, p<0.001, p<0.05, respectively). Neurological deficit score was remarkably decreased in the L. album group in comparison with the MCAO group (p<0.05).
Conclusions: It appears that pretreatment with L. album extract may attenuate brain tissue damage after ischemic stroke. The potential protective effects of this plant extract against this condition might be in part attributed to its anti-inflammatory and anti-apoptotic activities.
Collapse
|
85
|
Zhou M, Li R, Venkat P, Qian Y, Chopp M, Zacharek A, Landschoot-Ward J, Powell B, Jiang Q, Cui X. Post-Stroke Administration of L-4F Promotes Neurovascular and White Matter Remodeling in Type-2 Diabetic Stroke Mice. Front Neurol 2022; 13:863934. [PMID: 35572941 PMCID: PMC9100936 DOI: 10.3389/fneur.2022.863934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/21/2022] [Indexed: 02/02/2023] Open
Abstract
Patients with type 2 diabetes mellitus (T2DM) exhibit a distinct and high risk of ischemic stroke with worse post-stroke neurovascular and white matter (WM) prognosis than the non-diabetic population. In the central nervous system, the ATP-binding cassette transporter member A 1 (ABCA1), a reverse cholesterol transporter that efflux cellular cholesterol, plays an important role in high-density lipoprotein (HDL) biogenesis and in maintaining neurovascular stability and WM integrity. Our previous study shows that L-4F, an economical apolipoprotein A member I (ApoA-I) mimetic peptide, has neuroprotective effects via alleviating neurovascular and WM impairments in the brain of db/db-T2DM stroke mice. To further investigate whether L-4F has neurorestorative benefits in the ischemic brain after stroke in T2DM and elucidate the underlying molecular mechanisms, we subjected middle-aged, brain-ABCA1 deficient (ABCA1-B/-B), and ABCA1-floxed (ABCA1fl/fl) T2DM control mice to distal middle cerebral artery occlusion. L-4F (16 mg/kg, subcutaneous) treatment was initiated 24 h after stroke and administered once daily for 21 days. Treatment of T2DM-stroke with L-4F improved neurological functional outcome, and decreased hemorrhage, mortality, and BBB leakage identified by decreased albumin infiltration and increased tight-junction and astrocyte end-feet densities, increased cerebral arteriole diameter and smooth muscle cell number, and increased WM density and oligodendrogenesis in the ischemic brain in both ABCA1-B/-B and ABCA1fl/fl T2DM-stroke mice compared with vehicle-control mice, respectively (p < 0.05, n = 9 or 21/group). The L-4F treatment reduced macrophage infiltration and neuroinflammation identified by decreases in ED-1, monocyte chemoattractant protein-1 (MCP-1), and toll-like receptor 4 (TLR4) expression, and increases in anti-inflammatory factor Insulin-like growth factor 1 (IGF-1) and its receptor IGF-1 receptor β (IGF-1Rβ) in the ischemic brain (p < 0.05, n = 6/group). These results suggest that post-stroke administration of L-4F may provide a restorative strategy for T2DM-stroke by promoting neurovascular and WM remodeling. Reducing neuroinflammation in the injured brain may contribute at least partially to the restorative effects of L-4F independent of the ABCA1 signaling pathway.
Collapse
Affiliation(s)
- Min Zhou
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Rongwen Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Yu Qian
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Brianna Powell
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Quan Jiang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Xu Cui
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
86
|
Antioxidant Polyphenols of Antirhea borbonica Medicinal Plant and Caffeic Acid Reduce Cerebrovascular, Inflammatory and Metabolic Disorders Aggravated by High-Fat Diet-Induced Obesity in a Mouse Model of Stroke. Antioxidants (Basel) 2022; 11:antiox11050858. [PMID: 35624723 PMCID: PMC9138119 DOI: 10.3390/antiox11050858] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Metabolic disorders related to obesity and type 2 diabetes are associated with aggravated cerebrovascular damages during stroke. In particular, hyperglycemia alters redox and inflammatory status, leading to cerebral endothelial cell dysfunction, blood–brain barrier (BBB) disruption and brain homeostasis loss. Polyphenols constitute the most abundant dietary antioxidants and exert anti-inflammatory effects that may improve cerebrovascular complications in stroke. This study evaluated the effects of the characterized polyphenol-rich extract of Antirhea borbonica medicinal plant and its major constituent caffeic acid on a high-fat diet (HFD)-induced obesity mouse model during ischemic stroke, and murine bEnd3 cerebral endothelial cells in high glucose condition. In vivo, polyphenols administered by oral gavage for 12 weeks attenuated insulin resistance, hyperglycemia, hyperinsulinemia and dyslipidemia caused by HFD-induced obesity. Polyphenols limited brain infarct, hemorrhagic transformation and BBB disruption aggravated by obesity during stroke. Polyphenols exhibited anti-inflammatory and antioxidant properties by reducing IL-1β, IL-6, MCP-1, TNF-α and Nrf2 overproduction as well as total SOD activity elevation at the cerebral or peripheral levels in obese mice. In vitro, polyphenols decreased MMP-2 activity that correlated with MCP-1 secretion and ROS intracellular levels in hyperglycemic condition. Protective effects of polyphenols were linked to their bioavailability with evidence for circulating metabolites including caffeic acid, quercetin and hippuric acid. Altogether, these findings show that antioxidant polyphenols reduced cerebrovascular, inflammatory and metabolic disorders aggravated by obesity in a mouse model of stroke. It will be relevant to assess polyphenol-based strategies to improve the clinical consequences of stroke in the context of obesity and diabetes.
Collapse
|
87
|
Diaz A, Woo Y, Martin-Jimenez C, Merino P, Torre E, Yepes M. Tissue-type plasminogen activator induces TNF-α-mediated preconditioning of the blood-brain barrier. J Cereb Blood Flow Metab 2022; 42:667-682. [PMID: 34796748 PMCID: PMC9051146 DOI: 10.1177/0271678x211060395] [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: 05/10/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 11/15/2022]
Abstract
Ischemic tolerance is a phenomenon whereby transient exposure to a non-injurious preconditioning stimulus triggers resistance to a subsequent lethal ischemic insult. Despite the fact that not only neurons but also astrocytes and endothelial cells have a unique response to preconditioning stimuli, current research has been focused mostly on the effect of preconditioning on neuronal death. Thus, it is unclear if the blood-brain barrier (BBB) can be preconditioned independently of an effect on neuronal survival. The release of tissue-type plasminogen activator (tPA) from perivascular astrocytes in response to an ischemic insult increases the permeability of the BBB. In line with these observations, treatment with recombinant tPA increases the permeability of the BBB and genetic deficiency of tPA attenuates the development of post-ischemic edema. Here we show that tPA induces ischemic tolerance in the BBB independently of an effect on neuronal survival. We found that tPA renders the BBB resistant to an ischemic injury by inducing TNF-α-mediated astrocytic activation and increasing the abundance of aquaporin-4-immunoreactive astrocytic end-feet processes in the neurovascular unit. This is a new role for tPA, that does not require plasmin generation, and with potential therapeutic implications for patients with cerebrovascular disease.
Collapse
Affiliation(s)
- Ariel Diaz
- Division of Neuropharmacology and Neurologic Diseases, Yerkes
National Primate Research Center, Atlanta, GA, USA
| | - Yena Woo
- Division of Neuropharmacology and Neurologic Diseases, Yerkes
National Primate Research Center, Atlanta, GA, USA
| | - Cynthia Martin-Jimenez
- Division of Neuropharmacology and Neurologic Diseases, Yerkes
National Primate Research Center, Atlanta, GA, USA
| | - Paola Merino
- Division of Neuropharmacology and Neurologic Diseases, Yerkes
National Primate Research Center, Atlanta, GA, USA
| | - Enrique Torre
- Division of Neuropharmacology and Neurologic Diseases, Yerkes
National Primate Research Center, Atlanta, GA, USA
| | - Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Yerkes
National Primate Research Center, Atlanta, GA, USA
- Department of Neurology & Center for Neurodegenerative
Disease, Emory University, Atlanta, GA, USA
- Department of Neurology, Veterans Affairs Medical Center,
Atlanta, GA, USA
| |
Collapse
|
88
|
Tai SH, Huang SY, Chao LC, Lin YW, Huang CC, Wu TS, Shan YS, Lee AH, Lee EJ. Lithium upregulates growth-associated protein-43 (GAP-43) and postsynaptic density-95 (PSD-95) in cultured neurons exposed to oxygen-glucose deprivation and improves electrophysiological outcomes in rats subjected to transient focal cerebral ischemia following a long-term recovery period. Neurol Res 2022; 44:870-878. [PMID: 35348035 DOI: 10.1080/01616412.2022.2056817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Lithium has numerous neuroplastic and neuroprotective effects in patients with stroke. Here, we evaluated whether delayed and short-term lithium treatment reduces brain infarction volume and improves electrophysiological and neurobehavioral outcomes following long-term recovery after cerebral ischemia and the possible contributions of lithium-mediated mechanisms of neuroplasticity. METHODS Male Sprague Dawley rats were subjected to right middle cerebral artery occlusion for 90 min, followed by 28 days of recovery. Lithium chloride (1 mEq/kg) or vehicle was administered via intraperitoneal infusion once per day at 24 h after reperfusion onset. Neurobehavioral outcomes and somatosensory evoked potentials (SSEPs) were examined before and 28 days after ischemia-reperfusion. Brain infarction was assessed using Nissl staining. Primary cortical neuron cultures were exposed to oxygen-glucose deprivation (OGD) and treated with 2 or 20 μM lithium for 24 or 48 h; subsequent brain-derived neurotrophic factor (BDNF), growth-associated protein-43 (GAP-43), postsynaptic density-95 (PSD-95), and synaptosomal-associated protein-25 (SNAP-25) levels were analyzed using western blotting. RESULTS Compared to controls, lithium significantly reduced infarction volume in the ischemic brain and improved electrophysiological and neurobehavioral outcomes at 28 days post-insult. In cultured cortical neurons, BDNF, GAP-43, and PSD-95 expression were enhanced by 24- and 48-h treatment with lithium after OGD. CONCLUSION Lithium upregulates BDNF, GAP-43, and PSD-95, which partly accounts for its improvement of neuroplasticity and provision of long-term neuroprotection in the ischemic brain.Abbreviations: BDNF: brain-derived neurotrophic factor; ECM: extracellular matrix; EDTA: ethylenediaminetetraacetic acid; GAP-43: growth-associated protein-43; GSK-3β: glycogen synthase kinase-3β; HBSS: Hank's balanced salt solution; LCBF: local cortical blood perfusion; LDF: laser-Doppler flowmetry; MCAO: middle cerebral artery occlusion; MMP: matrix metalloproteinase; NMDA: N-methyl-D-aspartate; NMDAR: N-methyl-D-aspartate receptor; OCT: optimal cutting temperature compound; OGD: oxygen-glucose deprivation; PSD-95: postsynaptic density-95; SDS: sodium dodecyl sulfate; SNAP-25: synaptosomal-associated protein-25; SSEP: somatosensory evoked potential.
Collapse
Affiliation(s)
- Shih-Huang Tai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Yang Huang
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Liang-Chun Chao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Wen Lin
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Chih Huang
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tian-Shung Wu
- School of Pharmacy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ai-Hua Lee
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - E-Jian Lee
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
89
|
Chen X, Zhang J, Wu Y, Tucker R, Baird GL, Domonoske R, Barrios-Anderson A, Lim YP, Bath K, Walsh EG, Stonestreet BS. Inter-alpha Inhibitor Proteins Ameliorate Brain Injury and Improve Behavioral Outcomes in a Sex-Dependent Manner After Exposure to Neonatal Hypoxia Ischemia in Newborn and Young Adult Rats. Neurotherapeutics 2022; 19:528-549. [PMID: 35290609 PMCID: PMC9226254 DOI: 10.1007/s13311-022-01217-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/16/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is a major contributor to neurodevelopmental morbidities. Inter-alpha inhibitor proteins (IAIPs) have neuroprotective effects on HI-related brain injury in neonatal rats. However, the effects of treatment with IAIPs on sequential behavioral, MRI, and histopathological abnormalities in the young adult brain after treatment with IAIPs in neonates remain to be determined. The objective of this study was to examine the neuroprotective effects of IAIPs at different neurodevelopmental stages from newborn to young adults after exposure of neonates to HI injury. IAIPs were given as 11-sequential 30-mg/kg doses to postnatal (P) day 7-21 rats after right common carotid artery ligation and exposure to 90 min of 8% oxygen. The resulting brain edema and injury were examined by T2-weighted magnetic resonance imaging (MRI) and cresyl violet staining, respectively. The mean T2 values of the ipsilateral hemisphere from MRI slices 6 to 10 were reduced in IAIP-treated HI males + females on P8, P9, and P10 and females on P8, P9, P10, and P14. IAIP treatment reduced hemispheric volume atrophy by 44.5 ± 29.7% in adult male + female P42 rats and improved general locomotor abilities measured by the righting reflex over time at P7.5, P8, and P9 in males + females and males and muscle strength/endurance measured by wire hang on P16 in males + females and females. IAIPs provided beneficial effects during the learning phase of the Morris water maze with females exhibiting beneficial effects. IAIPs confer neuroprotection from HI-related brain injury in neonates and even in adult rats and beneficial MRI and behavioral benefits in a sex-dependent manner.
Collapse
Affiliation(s)
- Xiaodi Chen
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Jiyong Zhang
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Yuqi Wu
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Richard Tucker
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Grayson L Baird
- Department of Diagnostic Imaging, Biostatistics Core Lifespan Hospital System, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Rose Domonoske
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Adriel Barrios-Anderson
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin Bath
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical College, New York, NY, USA
| | - Edward G Walsh
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA.
| |
Collapse
|
90
|
Lohmann S, Grigoletto J, Bernis ME, Pesch V, Ma L, Reithofer S, Tamgüney G. Ischemic stroke causes Parkinson's disease-like pathology and symptoms in transgenic mice overexpressing alpha-synuclein. Acta Neuropathol Commun 2022; 10:26. [PMID: 35209932 PMCID: PMC8867857 DOI: 10.1186/s40478-022-01327-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/01/2022] [Indexed: 12/15/2022] Open
Abstract
The etiology of Parkinson's disease is poorly understood and is most commonly associated with advancing age, genetic predisposition, or environmental toxins. Epidemiological findings suggest that patients have a higher risk of developing Parkinson's disease after ischemic stroke, but this potential causality lacks mechanistic evidence. We investigated the long-term effects of ischemic stroke on pathogenesis in hemizygous TgM83 mice, which express human α-synuclein with the familial A53T mutation without developing any neuropathology or signs of neurologic disease for more than 600 days. We induced transient focal ischemia by middle cerebral artery occlusion in 2-month-old TgM83+/- mice and monitored their behavior and health status for up to 360 days post surgery. Groups of mice were sacrificed at 14, 30, 90, 180, and 360 days after surgery for neuropathological analysis of their brains. Motor deficits first appeared 6 months after focal ischemia and worsened until 12 months afterward. Immunohistochemical analysis revealed ischemia-induced neuronal loss in the infarct region and astrogliosis and microgliosis indicative of an inflammatory response, which was most pronounced at 14 days post surgery. Infarct volume and inflammation gradually decreased in size and severity until 180 days post surgery. Surprisingly, neuronal loss and inflammation were increased again by 360 days post surgery. These changes were accompanied by a continuous increase in α-synuclein aggregation, its neuronal deposition, and a late loss of dopaminergic neurons in the substantia nigra, which we detected at 360 days post surgery. Control animals that underwent sham surgery without middle cerebral artery occlusion showed no signs of disease or neuropathology. Our results establish a mechanistic link between ischemic stroke and Parkinson's disease and provide an animal model for studying possible interventions.
Collapse
Affiliation(s)
- Stephanie Lohmann
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Jessica Grigoletto
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Maria Eugenia Bernis
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Verena Pesch
- Institut für Biologische Informationsprozesse, Strukturbiochemie (IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425, Jülich, Germany
| | - Liang Ma
- Institut für Biologische Informationsprozesse, Strukturbiochemie (IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425, Jülich, Germany
| | - Sara Reithofer
- Institut für Biologische Informationsprozesse, Strukturbiochemie (IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425, Jülich, Germany
| | - Gültekin Tamgüney
- Institut für Biologische Informationsprozesse, Strukturbiochemie (IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425, Jülich, Germany.
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
91
|
Li L, Yasmen N, Hou R, Yang S, Lee JY, Hao J, Yu Y, Jiang J. Inducible Prostaglandin E Synthase as a Pharmacological Target for Ischemic Stroke. Neurotherapeutics 2022; 19:366-385. [PMID: 35099767 PMCID: PMC9130433 DOI: 10.1007/s13311-022-01191-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2022] [Indexed: 01/03/2023] Open
Abstract
As the inducible terminal enzyme for prostaglandin E2 (PGE2) synthesis, microsomal PGE synthase-1 (mPGES-1) contributes to neuroinflammation and secondary brain injury after cerebral ischemia via producing excessive PGE2. However, a proof of concept that mPGES-1 is a therapeutic target for ischemic stroke has not been established by a pharmacological strategy mainly due to the lack of drug-like mPGES-1 inhibitors that can be used in relevant rodent models. To this end, we recently developed a series of novel small-molecule compounds that can inhibit both human and rodent mPGES-1. In this study, blockade of mPGES-1 by our several novel compounds abolished the lipopolysaccharide (LPS)-induced PGE2 and pro-inflammatory cytokines interleukin 1β (IL-1β), IL-6, and tumor necrosis factor α (TNF-α) in mouse primary brain microglia. Inhibition of mPGES-1 also decreased PGE2 produced by neuronal cells under oxygen-glucose deprivation (OGD) stress. Among the five enzymes for PGE2 biosynthesis, mPGES-1 was the most induced one in cerebral ischemic lesions. Systemic treatment with our lead compound MPO-0063 (5 or 10 mg/kg, i.p.) in mice after transient middle cerebral artery occlusion (MCAO) improved post-stroke well-being, decreased infarction and edema, suppressed induction of brain cytokines (IL-1β, IL-6, and TNF-α), alleviated locomotor dysfunction and anxiety-like behavior, and reduced the long-term cognitive impairments. The therapeutic effects of MPO-0063 in this proof-of-concept study provide the first pharmacological evidence that mPGES-1 represents a feasible target for delayed, adjunct treatment - along with reperfusion therapies - for acute brain ischemia.
Collapse
Affiliation(s)
- Lexiao Li
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Nelufar Yasmen
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Ruida Hou
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Seyoung Yang
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jae Yeol Lee
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jiukuan Hao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204, USA
| | - Ying Yu
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
92
|
ADAM8 Activates NLRP3 Inflammasome to Promote Cerebral Ischemia-Reperfusion Injury. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:3097432. [PMID: 34956566 PMCID: PMC8702307 DOI: 10.1155/2021/3097432] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/03/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Stroke is the leading cause of death and disability in humans. Strokes are classified as either ischemic or hemorrhagic. Ischemic stroke accounts for 70–80% of the cases. Inflammation is a key factor in ischemic brain injury. Studies have shown that inflammatory response induced by NLRP3 inflammasome is one of the root causes of brain damage in mice with cerebral ischemia. However, its specific mechanism in cerebral ischemia is still unclear. ADAM8 (a disintegrin and metalloproteases 8) is a transmembrane protein with different functions. It plays an important role in tumors and neuroinflammation-related diseases. However, the role and molecular mechanism of ADAM8 in cerebral ischemia injury are still unclear. This study aims to evaluate the role of ADAM8 in cerebral ischemic injury and explore its signal transduction mechanism. This experiment shows that ADAM8 can significantly cause neurological deficits in MCAO mice and can substantially cause ipsilateral cerebral edema and cerebral infarction in MCAO mice. In addition, ADAM8 can significantly induce cortical cell apoptosis in MCAO mice, leading to the loss of neurons and the expression of proinflammatory factors COX2, iNOS, TNFα, and IL-6. Importantly, we confirmed that ADAM8 mediates the inflammatory response by promoting the activation of NLRP3 inflammasome, microglia, and astrocytes. These results indicate that ADAM8 may be a candidate drug target for the prevention and treatment of the cerebral ischemic injury.
Collapse
|
93
|
Lai AKW, Ng TC, Hung VKL, Tam KC, Cheung CW, Chung SK, Lo ACY. Exacerbated VEGF up-regulation accompanies diabetes-aggravated hemorrhage in mice after experimental cerebral ischemia and delayed reperfusion. Neural Regen Res 2021; 17:1566-1575. [PMID: 34916442 PMCID: PMC8771109 DOI: 10.4103/1673-5374.330612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Reperfusion therapy is the preferred treatment for ischemic stroke, but is hindered by its short treatment window, especially in patients with diabetes whose reperfusion after prolonged ischemia is often accompanied by exacerbated hemorrhage. The mechanisms underlying exacerbated hemorrhage are not fully understood. This study aimed to identify this mechanism by inducing prolonged 2-hour transient intraluminal middle cerebral artery occlusion in diabetic Ins2Akita/+ mice to mimic patients with diabetes undergoing delayed mechanical thrombectomy. The results showed that at as early as 2 hours after reperfusion, Ins2Akita/+ mice exhibited rapid development of neurological deficits, increased infarct and hemorrhagic transformation, together with exacerbated down-regulation of tight-junction protein ZO-1 and up-regulation of blood-brain barrier-disrupting matrix metallopeptidase 2 and matrix metallopeptidase 9 when compared with normoglycemic Ins2+/+ mice. This indicated that diabetes led to the rapid compromise of vessel integrity immediately after reperfusion, and consequently earlier death and further aggravation of hemorrhagic transformation 22 hours after reperfusion. This observation was associated with earlier and stronger up-regulation of pro-angiogenic vascular endothelial growth factor (VEGF) and its downstream phospho-Erk1/2 at 2 hours after reperfusion, which was suggestive of premature angiogenesis induced by early VEGF up-regulation, resulting in rapid vessel disintegration in diabetic stroke. Endoplasmic reticulum stress-related pro-apoptotic C/EBP homologous protein was overexpressed in challenged Ins2Akita/+ mice, which suggests that the exacerbated VEGF up-regulation may be caused by overwhelming endoplasmic reticulum stress under diabetic conditions. In conclusion, the results mimicked complications in patients with diabetes undergoing delayed mechanical thrombectomy, and diabetes-induced accelerated VEGF up-regulation is likely to underlie exacerbated hemorrhagic transformation. Thus, suppression of the VEGF pathway could be a potential approach to allow reperfusion therapy in patients with diabetic stroke beyond the current treatment window. Experiments were approved by the Committee on the Use of Live Animals in Teaching and Research of the University of Hong Kong [CULATR 3834-15 (approval date January 5, 2016); 3977-16 (approval date April 13, 2016); and 4666-18 (approval date March 29, 2018)].
Collapse
Affiliation(s)
- Angela Ka Wai Lai
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Tsz Chung Ng
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Victor Ka Lok Hung
- Department of Anesthesiology, Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Ka Cheung Tam
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Chi Wai Cheung
- Department of Anesthesiology, Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Sookja Kim Chung
- Macau University of Science and Technology, Taipa, Macau Special Administration Region; School of Biomedical Sciences, The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Amy Cheuk Yin Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| |
Collapse
|
94
|
Wang LC, Wei WY, Ho PC, Wu PY, Chu YP, Tsai KJ. Somatosensory Cortical Electrical Stimulation After Reperfusion Attenuates Ischemia/Reperfusion Injury of Rat Brain. Front Aging Neurosci 2021; 13:741168. [PMID: 34867274 PMCID: PMC8632773 DOI: 10.3389/fnagi.2021.741168] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/11/2021] [Indexed: 01/01/2023] Open
Abstract
Objective: Ischemic stroke is an important cause of death and disability worldwide. Early reperfusion by thrombolysis or thrombectomy has improved the outcome of acute ischemic stroke. However, the therapeutic window for reperfusion therapy is narrow, and adjuvant therapy for neuroprotection is demanded. Electrical stimulation (ES) has been reported to be neuroprotective in many neurological diseases. In this study, the neuroprotective effect of early somatosensory cortical ES in the acute stage of ischemia/reperfusion injury was evaluated. Methods: In this study, the rat model of transient middle cerebral artery occlusion was used to explore the neuroprotective effect and underlying mechanisms of direct primary somatosensory (S1) cortex ES with an electric current of 20 Hz, 2 ms biphasic pulse, 100 μA for 30 min, starting at 30 min after reperfusion. Results: These results showed that S1 cortical ES after reperfusion decreased infarction volume and improved functional outcome. The number of activated microglia, astrocytes, and cleaved caspase-3 positive neurons after ischemia/reperfusion injury were reduced, demonstrating that S1 cortical ES alleviates inflammation and apoptosis. Brain-derived neurotrophic factor (BDNF) and phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway were upregulated in the penumbra area, suggesting that BDNF/TrkB signals and their downstream PI3K/Akt signaling pathway play roles in ES-related neuroprotection. Conclusion: This study demonstrates that somatosensory cortical ES soon after reperfusion can attenuate ischemia/reperfusion injury and is a promising adjuvant therapy for thrombolytic treatment after acute ischemic stroke. Advanced techniques and devices for high-definition transcranial direct current stimulation still deserve further development in this regard.
Collapse
Affiliation(s)
- Liang-Chao Wang
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Yen Wei
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chuan Ho
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Yi Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Ping Chu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
95
|
Zhou M, Li D, Li L, Zhao P, Yue S, Li X, Du Y, Fan X, Zhang M. Post-stroke treatment of storax improves long-term outcomes of stroke in rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114467. [PMID: 34343648 DOI: 10.1016/j.jep.2021.114467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 06/28/2021] [Accepted: 07/25/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The lack of widely applicable pharmacological treatments for ischemic stroke patients has led to a growing interest in traditional medicine. The identification of pharmacologically active components of the clinically used traditional medicine has been considered translationally significant. SuHeXiang Wan is a prescription containing 15 herbs approved by Chinese National Drug Administration (CNDA) for treating ischemic stroke. Storax is one of the main contents in this prescription and is believed to play a significant pharmacological role, which has been used to treat stroke for nearly 1000 years in traditional Chinese medicine. Emerging experimental investigations suggested Storax might be effective for treating ischemic stroke. AIM OF THE STUDY This study aimed to test our hypothesis that post-stroke treatment of Storax can improve long-term outcomes of stroke. MATERIALS AND METHODS Male Wistar rats (250-280 g) subjected to 2 h of MCAO following reperfusion were treated with Storax by intragastric at 1 h and repeated at 3 h, 6 h after stroke. In the first set experiment, an array of neurological function deficits assessments were tested before and after stroke, brain lesion size was examined at 28 days after ischemia. CD31 and synatophysin were analyzed by immunohistochemistry. In the second set experiment, markers of proinflammatory activation were determined at 24 h after stroke. ELISA was performed to analyze brain concentrations of TNF-α, IL-1β and circulating levels of iNOS, ET-1, and immunohistochemistry was performed to determine GFAP, IBA-1 and NF-κB p65. RESULTS Storax significantly alleviated neurological deficits from 7 days after stroke and lasted until 28 days, corresponding to the significantly decreased lesion volume at 28 days after stroke; Meanwhile, Storax increased the density of CD31and SYP in peri-infarct areas. At 24 h after stroke, Storax significantly inhibited brain TNF-α, IL-1β expression and circulating iNOS, ET-1 levels, reduced the NF-κB/p65 positive cell number, and decreased activated microglia/macrophages and astrocytes cell numbers alongside reversed their morphological transformations. CONCLUSION Our experimental findings demonstrate treatment of Storax at the acute phase significantly improves long-term neurological outcomes in the focal stroke model of rats. We also speculate that inhibition of acute proinflammation activation by Storax might be associated with its beneficial pharmacological effect, but remain to define and elucidate in future investigation.
Collapse
Affiliation(s)
- Min Zhou
- Department of Traditional Chinese Medicine, Tianjin Medical University General Hospital, Tianjin, China.
| | - Dongna Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Pei Zhao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Shaoqian Yue
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Yuzheng Du
- Acupuncture and Moxibustion Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xiang Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Meng Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
96
|
Bonetti NR, Meister TA, Soria R, Akhmedov A, Liberale L, Ministrini S, Dogar A, Lüscher TF, Messerli FH, Rexhaj E, Camici GG, Beer JH, Scherrer U. In vitro fertilization exacerbates stroke size and neurological disability in wildtype mice. Int J Cardiol 2021; 343:92-101. [PMID: 34437933 DOI: 10.1016/j.ijcard.2021.08.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/28/2021] [Accepted: 08/19/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND PURPOSE Assisted reproductive technologies (ART) induce premature vascular aging in human offspring. The related alterations are well-established risk factors for stroke and predictors of adverse stroke outcome. However, given the young age of the human ART population there is no information on the incidence and outcome of cerebrovascular complications in humans. In mice, ART alters the cardiovascular phenotype similarly to humans, thereby offering the possibility to study this problem. METHODS We investigated the morphological and clinical outcome after ischemia/reperfusion brain injury induced by transient (45 min) middle cerebral artery occlusion in ART and control mice. RESULTS We found that stroke volumes were almost 3-fold larger in ART than in control mice (P < 0.001). In line with these morphological differences, neurological performance assessed by the Bederson and RotaRod tests 24 and 48 h after artery occlusion was significantly worse in ART compared with control mice. Plasma levels of TNF-alpha, were also significantly increased in ART vs. control mice after stroke (P < 0.05). As potential underlying mechanisms, we identified increased blood-brain barrier permeability evidenced by increased IgG extravasation associated with decreased tight junctional protein claudin-5 and occludin expression, increased oxidative stress and decreased NO-bioactivity in ART compared with control mice. CONCLUSIONS In wildtype mice, ART predisposes to significantly worse morphological and functional stroke outcomes, related at least in part to altered blood-brain barrier permeability. These findings demonstrate that ART, by inducing premature vascular aging, not only is a likely risk factor for stroke-occurrence, but also a mediator of adverse stroke-outcome. TRANSLATIONAL PERSPECTIVE This study highlights that ART not only is a likely risk factor for stroke-occurrence, but also a mediator of adverse stroke-outcome. The findings should raise awareness in the ever-growing human ART population in whom these techniques cause similar alterations of the cardiovascular phenotype and encourage early preventive and diagnostic efforts.
Collapse
Affiliation(s)
- N R Bonetti
- Center for Molecular Cardiology, University of Zurich, Switzerland; Department of Internal Medicine, Kantonsspital Baden, Baden, Switzerland
| | - T A Meister
- Department of Cardiology and Biomedical Research, Inselspital, University of Bern, Switzerland
| | - R Soria
- Department of Cardiology and Biomedical Research, Inselspital, University of Bern, Switzerland
| | - A Akhmedov
- Center for Molecular Cardiology, University of Zurich, Switzerland
| | - L Liberale
- Center for Molecular Cardiology, University of Zurich, Switzerland; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - S Ministrini
- Center for Molecular Cardiology, University of Zurich, Switzerland; Department of Medicine and Surgery, Università degli Studi di Perugia, Perugia, Italy
| | - A Dogar
- Department of Cardiology and Biomedical Research, Inselspital, University of Bern, Switzerland; Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore 54792, Pakistan
| | - T F Lüscher
- Center for Molecular Cardiology, University of Zurich, Switzerland; Royal Brompton and Harefield Hospital Trusts, London, UK
| | - F H Messerli
- Department of Cardiology and Biomedical Research, Inselspital, University of Bern, Switzerland
| | - E Rexhaj
- Department of Cardiology and Biomedical Research, Inselspital, University of Bern, Switzerland
| | - G G Camici
- Center for Molecular Cardiology, University of Zurich, Switzerland; University Heart Center, University Hospital Zurich, Switzerland; Department of Research and Education, University Hospital Zurich, Switzerland
| | - J H Beer
- Center for Molecular Cardiology, University of Zurich, Switzerland; Department of Internal Medicine, Kantonsspital Baden, Baden, Switzerland
| | - U Scherrer
- Department of Cardiology and Biomedical Research, Inselspital, University of Bern, Switzerland; Facultad de Ciencias, Departamento de Biología, Universidad de Tarapacá, Arica, Chile.
| |
Collapse
|
97
|
Ahmad N, Mishra A, Ahsan F, Khan Z. Evaluation of cerebroprotective effect of Ricinus communis leaves against ischemia reperfusion injury in rats. CLINICAL PHYTOSCIENCE 2021. [DOI: 10.1186/s40816-021-00323-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Ricinus communis (RC) has been used for a long time as natural origin medicine in the treatment of central nervous system ailments. This present study was designed to identify the possible role of Ricinus communis leaves extract against ischemia-reperfusion induced-neurobehavioral changes, oxidative stress, histopathological and cellular modifications in the brain.
Methods
Sprague Dawley (SD) rats (200–250 g) were induced to bilateral common carotid artery occlusion (BCCAO) for around 30 min later subjected to reperfusion for 24 h to induce cerebral injury by reperfusion. Ricinus communis leaves extract (250 and 500 mg/kg, p.o) was administered continuously for 14 days and on the 15th-day animals were subjected to ischemia-reperfusion injury. Different behavioral tests and biochemical parameters were assessed subsequently.
Results
Fourteen days Ricinus communis leaves extract (250 and 500 mg/kg, p.o.) treatment very significantly improved neurobehavioral alterations when compared to control ischemia-reperfusion. Ricinus communis leaves extract (250 and 500 mg/kg, p.o.) kg, i.p. treatment significantly attenuated oxidative damage when compared to ischemia-reperfusion (I/R) group animals. In addition, Ricinus communis leaves extract treatment was well supported histopathologically when compared to the ischemia-reperfusion (I/R) group.
Conclusion
The data from this study recommend that treatment with Ricinus communis leaves extract increases the antioxidant protection against BCCAO-induced global cerebral ischemia and demonstrates neuroprotective activity.
Collapse
|
98
|
Mi Z, Liu H, Rose ME, Ma J, Reay DP, Ma X, Henchir JJ, Dixon CE, Graham SH. Mutation of a Ubiquitin Carboxy Terminal Hydrolase L1 Lipid Binding Site Alleviates Cell Death, Axonal Injury, and Behavioral Deficits After Traumatic Brain Injury in Mice. Neuroscience 2021; 475:127-136. [PMID: 34508847 DOI: 10.1016/j.neuroscience.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022]
Abstract
Ubiquitin carboxy terminal hydrolase L1 (UCHL1) is a protein highly expressed in neurons that may play important roles in the ubiquitin proteasome pathway (UPP) in neurons, axonal integrity, and motor function after traumatic brain injury (TBI). Binding of reactive lipid species to cysteine 152 of UCHL1 results in unfolding, aggregation, and inactivation of the enzyme. To test the role of this mechanism in TBI, mice bearing a cysteine to alanine mutation at site 152 (C152A mice) that renders UCHL1 resistant to inactivation by reactive lipids were subjected to the controlled cortical impact model (CCI) of TBI and compared to wild type (WT) controls. Alterations in protein ubiquitination and activation of autophagy pathway markers in traumatized brain were detected by immunoblotting. Cell death and axonal injury were determined by histological assessment and anti-amyloid precursor protein (APP) immunohistochemistry. Behavioral outcomes were determined using the beam balance and Morris water maze tests. C152A mice had reduced accumulation of ubiquitinated proteins, decreased activation of the autophagy markers Beclin-1 and LC3B, a decreased number of abnormal axons, decreased CA1 cell death, and improved motor and cognitive function compared to WT controls after CCI; no significant change in spared tissue volume was observed. These results suggest that binding of lipid substrates to cysteine 152 of UCHL1 is important in the pathogenesis of injury and recovery after TBI and may be a novel target for future therapeutic approaches.
Collapse
Affiliation(s)
- Zhiping Mi
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Hao Liu
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA
| | - Marie E Rose
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Jie Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Daniel P Reay
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Xiecheng Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Jeremy J Henchir
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | - C Edward Dixon
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | - Steven H Graham
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
99
|
Yu-lan T, Mai-lan L, Jian L, Nan L, Guo-shan Z, Jie Y, Qian-yun Y. Exploring the effect of acupuncture plus mild hypothermia on miRNA-204 and its target gene expressions in CIRI rat brain tissues based on MAPK signal pathway. JOURNAL OF ACUPUNCTURE AND TUINA SCIENCE 2021. [DOI: 10.1007/s11726-021-1264-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
100
|
Hao S, Zhong Z, Qu W, Huang Z, Sun F, Qiu M. Melatonin supplementation in the subacute phase after ischemia alleviates postischemic sleep disturbances in rats. Brain Behav 2021; 11:e2366. [PMID: 34520636 PMCID: PMC8553311 DOI: 10.1002/brb3.2366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/25/2021] [Accepted: 08/31/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Sleep disorders are highly prevalent among stroke survivors and impede stroke recovery. It is well established that melatonin has neuroprotective effects in animal models of ischemic stroke. However, as a modulator of endogenous physiological circadian rhythms, the effects of melatonin on poststroke sleep disorders remain unclear. In the present study, we investigated how melatonin delivered intraperitoneally once daily in the subacute phase after stroke onset, influencing neuronal survival, motor recovery, and sleep-wake profiles in rats. METHODS Transient ischemic stroke in male Sprague-Dawley rats was induced with 30 min occlusion of the middle cerebral artery. Melatonin or vehicle was delivered intraperitoneally once daily in the subacute phase, from 2 to 7 days after stroke. Electroencephalogram and electromyogram recordings were obtained simultaneously. RESULTS Compared to the effects observed in the vehicle-treated ischemic group, after 6 daily consecutive treatment of melatonin at 10 mg/kg starting at ischemic/reperfusion day 2, the infarct volume was significantly decreased (from 39.6 to 26.2%), and the degeneration of axons in the ipsilateral striatum and the contralateral corpus callosum were significantly alleviated. Sensorimotor performances were obviously improved as evidenced by significant increases in the latency to falling off the wire and in the use of the impaired forelimb. In addition to those predictable results of reducing brain tissue damage and mitigating behavioral deficits, repeated melatonin treatment during the subacute phase of stroke also alleviated sleep fragmentation through reducing sleep-wake stage transitions and stage bouts, together with increasing stage durations. Furthermore, daily administration of melatonin at 9 a.m. significantly increased the nonrapid eye movement sleep delta power during both the light and dark periods and decreased the degree of reduction of the circadian index. CONCLUSIONS Melatonin promptly reversed ischemia-induced sleep disturbances. The neuroprotective effects of melatonin on ischemic injury may be partially associated with its role in sleep modulation.
Collapse
Affiliation(s)
- Shu‐Mei Hao
- Department of NeurobiologyInstitute for Basic Research on Aging and MedicineSchool of Basic Medical ScienceFudan UniversityShanghaiChina
| | - Zhi‐Gang Zhong
- Department of NeurobiologyInstitute for Basic Research on Aging and MedicineSchool of Basic Medical ScienceFudan UniversityShanghaiChina
- Department of PharmacologySchool of Basic Medical ScienceState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Wei‐Min Qu
- Department of PharmacologySchool of Basic Medical ScienceState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Zhi‐Li Huang
- Department of PharmacologySchool of Basic Medical ScienceState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Feng‐Yan Sun
- Department of NeurobiologyInstitute for Basic Research on Aging and MedicineSchool of Basic Medical ScienceFudan UniversityShanghaiChina
| | - Mei‐Hong Qiu
- Department of NeurobiologyInstitute for Basic Research on Aging and MedicineSchool of Basic Medical ScienceFudan UniversityShanghaiChina
- Department of PharmacologySchool of Basic Medical ScienceState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|