1
|
Tsuruta K, Fukushima H, Sakai H. Hemoglobin vesicles improve neurological outcomes after cardiac arrest in rats. Resusc Plus 2024; 20:100819. [PMID: 39559730 PMCID: PMC11570973 DOI: 10.1016/j.resplu.2024.100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
Aim To investigate the effects of hemoglobin vesicles (HbVs) in preventing hypoxic brain injury after cardiac arrest in a rat model of asphyxia-related cardiac arrest. Methods Male Wistar rats were divided into three groups: HbVs (n = 18), control (n = 29), and sham (n = 7). Respiratory arrest was induced using muscle relaxants under ventilation. Cardiac arrest occurred 3-4 min later. After 8 min, HbVs or saline (5 ml/kg), adrenaline, and sodium bicarbonate were administered, followed by chest compressions and ventilation. Resuscitation was deemed successful with a mean arterial pressure > 60 mmHg sustained for at least 5 min. Behavioral and histopathological evaluations were performed 7 days later. Results Survival rates were 39 % and 24 % in the HbVs and control groups, respectively (P = 0.308). Motor activity scores and spatial memory were significantly higher in the HbVs group (P < 0.001). Hippocampal CA1 region staining indicated significantly less neuropathy in the HbVs group (P < 0.001). Conclusion The administration of HbVs during resuscitation was effective in mitigating brain damage after whole-brain ischemia in rats, as demonstrated by improved histopathological and neurological outcomes. This suggests potential neurological benefits for patients during resuscitation, although further research in larger animal models is required to validate these findings.
Collapse
Affiliation(s)
- Keisuke Tsuruta
- Department of Emergency and Critical Care Medicine, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
- Department of Chemistry, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Hidetada Fukushima
- Department of Emergency and Critical Care Medicine, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Hiromi Sakai
- Department of Chemistry, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| |
Collapse
|
2
|
Wang H, Li J, Wu G, Lin X, Chen J, Liang J, Zhang J, Luo X, Mao H, Xie J, Li Z, Zhou H, Xu K, Yin J, He Y. Activated sympathetic nerve post stroke downregulates Toll-like receptor 5 and disrupts the gut mucosal barrier. Cell Rep Med 2024; 5:101754. [PMID: 39383869 PMCID: PMC11513850 DOI: 10.1016/j.xcrm.2024.101754] [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: 12/19/2023] [Revised: 07/27/2024] [Accepted: 09/06/2024] [Indexed: 10/11/2024]
Abstract
The gut permeability significantly increases after ischemic stroke, partly due to disrupted mucosal barrier, but the mechanism remains elusive. Here, we found that the mucus disruption starts at 2 h post stroke, whereas goblet cell functions remain intact. Meanwhile, the flagellated bacteria Helicobacter thrives and penetrates in the mucus layer. Elimination of the mucosal microbiota or transplantation of Helicobacter in germ-free mice reveals an important role of the mucosal microbiota in mucus disruption. The bacterial invasion is due to downregulated Toll-like receptor 5 (TLR5) and its downstream products flagellin-specific IgA and antimicrobial peptides. Knockdown of intestinal TLR5 increases the abundance of flagellated bacteria and exacerbates mucus injury. Intestinal TLR5 is downregulated by the activation of sympathetic nerve. Serum noradrenaline level is positively associated with flagellin level in patients with stroke and patients' prognosis. These findings reveal a neural pathway in which the sympathetic nerve disrupts the mucosal barrier, providing potential therapeutic targets for stroke injury.
Collapse
Affiliation(s)
- Huidi Wang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jie Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Guangyan Wu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xiaofei Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiaying Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jingru Liang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiahui Zhang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xiaoxia Luo
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Hongyun Mao
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jiahui Xie
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhuang Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong 510033, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Kaiyu Xu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China.
| | - Jia Yin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong 510033, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
3
|
Koukalova L, Chmelova M, Amlerova Z, Vargova L. Out of the core: the impact of focal ischemia in regions beyond the penumbra. Front Cell Neurosci 2024; 18:1336886. [PMID: 38504666 PMCID: PMC10948541 DOI: 10.3389/fncel.2024.1336886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/08/2024] [Indexed: 03/21/2024] Open
Abstract
The changes in the necrotic core and the penumbra following induction of focal ischemia have been the focus of attention for some time. However, evidence shows, that ischemic injury is not confined to the primarily affected structures and may influence the remote areas as well. Yet many studies fail to probe into the structures beyond the penumbra, and possibly do not even find any significant results due to their short-term design, as secondary damage occurs later. This slower reaction can be perceived as a therapeutic opportunity, in contrast to the ischemic core defined as irreversibly damaged tissue, where the window for salvation is comparatively short. The pathologies in remote structures occur relatively frequently and are clearly linked to the post-stroke neurological outcome. In order to develop efficient therapies, a deeper understanding of what exactly happens in the exo-focal regions is necessary. The mechanisms of glia contribution to the ischemic damage in core/penumbra are relatively well described and include impaired ion homeostasis, excessive cell swelling, glutamate excitotoxic mechanism, release of pro-inflammatory cytokines and phagocytosis or damage propagation via astrocytic syncytia. However, little is known about glia involvement in post-ischemic processes in remote areas. In this literature review, we discuss the definitions of the terms "ischemic core", "penumbra" and "remote areas." Furthermore, we present evidence showing the array of structural and functional changes in the more remote regions from the primary site of focal ischemia, with a special focus on glia and the extracellular matrix. The collected information is compared with the processes commonly occurring in the ischemic core or in the penumbra. Moreover, the possible causes of this phenomenon and the approaches for investigation are described, and finally, we evaluate the efficacy of therapies, which have been studied for their anti-ischemic effect in remote areas in recent years.
Collapse
Affiliation(s)
- Ludmila Koukalova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
4
|
Kapasi A, Capuano AW, Lamar M, Leurgans SE, Evia AM, Bennett DA, Arfanakis K, Schneider JA. Atherosclerosis and Hippocampal Volumes in Older Adults: The Role of Age and Blood Pressure. J Am Heart Assoc 2024; 13:e031551. [PMID: 38240240 PMCID: PMC11056126 DOI: 10.1161/jaha.123.031551] [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: 09/13/2023] [Accepted: 12/05/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Lower hippocampal volume is associated with late-life cognitive decline and is an important, but nonspecific marker for clinical Alzheimer's dementia. Cerebrovascular disease may also be associated with hippocampal volume. Here we study the role of intracranial large vessel disease (atherosclerosis) in association with hippocampal volume and the potential role of age, average late-life blood pressure across all visits, and other factors (sex, apolipoprotein ε4 [APOE ε4], and diabetes). METHODS AND RESULTS Data came from 765 community-based older people (91 years old on average at death; 72% women), from 2 ongoing clinical-pathologic cohort studies. Participants completed baseline assessment, annual standardized blood pressure measurements, vascular risk assessment for diabetes, and blood draws to determine APOE genotype, and at death, brains were removed and underwent ex vivo magnetic resonance imaging and neuropathologic evaluation for atherosclerosis pathology and other cerebrovascular and neurodegenerative pathologies. Linear regression models examined the association of atherosclerosis and hippocampal to hemisphere volume ratio and whether age at death, blood pressure, and other factors modified associations. In linear regression models adjusted for demographics and neurodegenerative and other cerebrovascular pathologies, atherosclerosis severity was associated with a lower hippocampal to hemisphere volume ratio. In separate models, we found the effect of atherosclerosis on the ratio of hippocampal to hemisphere volume was attenuated among advanced age at death or having higher systolic blood pressure (interaction terms P≤0.03). We did not find confounding or interactions with sex, diabetes, or APOE ε4. CONCLUSIONS Atherosclerosis severity is associated with lower hippocampal volume, independent of neurodegenerative and other cerebrovascular pathologies. Higher systolic blood pressures and advanced age attenuate associations.
Collapse
Affiliation(s)
- Alifiya Kapasi
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
- Department of Pathology (Neuropathology)Rush University Medical CenterChicagoIL
| | - Ana W. Capuano
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
- Department of Neurological SciencesRush University Medical CenterChicagoIL
| | - Melissa Lamar
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
- Department of Psychiatry and Behavioral SciencesRush University Medical CenterChicagoIL
| | - Sue E. Leurgans
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
- Department of Neurological SciencesRush University Medical CenterChicagoIL
| | - Arnold M. Evia
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
| | - David A. Bennett
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
- Department of Neurological SciencesRush University Medical CenterChicagoIL
| | - Konstantinos Arfanakis
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
- Department of Biomedical EngineeringIllinois Institute of TechnologyChicagoIL
- Department of Diagnostic RadiologyRush University Medical CenterChicagoIL
| | - Julie A. Schneider
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
- Department of Pathology (Neuropathology)Rush University Medical CenterChicagoIL
- Department of Neurological SciencesRush University Medical CenterChicagoIL
| |
Collapse
|
5
|
Gannon O, Tremble SM, McGinn C, Guth R, Scoppettone N, Hunt RD, Prakash K, Johnson AC. Angiotensin II-mediated hippocampal hypoperfusion and vascular dysfunction contribute to vascular cognitive impairment in aged hypertensive rats. Alzheimers Dement 2024; 20:890-903. [PMID: 37817376 PMCID: PMC10917018 DOI: 10.1002/alz.13491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Chronic hypertension increases the risk of vascular cognitive impairment (VCI) by ∼60%; however, how hypertension affects the vasculature of the hippocampus remains unclear but could contribute to VCI. METHODS Memory, hippocampal perfusion, and hippocampal arteriole (HA) function were investigated in male Wistar rats or spontaneously hypertensive rats (SHR) in early (4 to 5 months old), mid (8 to 9 months old), or late adulthood (14 to 15 months old). SHR in late adulthood were chronically treated with captopril (angiotensin converting enzyme inhibitor) or apocynin (antioxidant) to investigate the mechanisms by which hypertension contributes to VCI. RESULTS Impaired memory in SHR in late adulthood was associated with HA endothelial dysfunction, hyperconstriction, and ∼50% reduction in hippocampal blood flow. Captopril, but not apocynin, improved HA function, restored perfusion, and rescued memory function in aged SHR. DISCUSSION Hippocampal vascular dysfunction contributes to hypertension-induced memory decline through angiotensin II signaling, highlighting the therapeutic potential of HAs in protecting neurocognitive health later in life. HIGHLIGHTS Vascular dysfunction in the hippocampus contributes to vascular cognitive impairment. Memory declines with age during chronic hypertension. Angiotensin II causes endothelial dysfunction in the hippocampus in hypertension. Angiotensin II-mediated hippocampal arteriole dysfunction reduces blood flow. Vascular dysfunction in the hippocampus impairs perfusion and memory function.
Collapse
Affiliation(s)
- Olivia Gannon
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Sarah M. Tremble
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Conor McGinn
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Ruby Guth
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Nadia Scoppettone
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Ryan D. Hunt
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Kirtika Prakash
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Abbie C. Johnson
- Department of Neurological SciencesUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| |
Collapse
|
6
|
Ghatak S, Kumar Sikdar S. Prolonged exposure to lactate causes TREK1 channel clustering in rat hippocampal astrocytes. Neurosci Lett 2024; 821:137613. [PMID: 38157928 DOI: 10.1016/j.neulet.2023.137613] [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: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Increased concentrations of lactate (15-30 mM) are associated with and found to be neuroprotective in various brain pathophysiology. In our earlier studies we showed that high levels of lactate can increase TREK1 channel activity and expression within 1 h. TREK1 channels are two pore domain leak potassium ion channels that are upregulated during cerebral ischemia, epilepsy and other brain pathologies. They play a prominent neuroprotective role against excitotoxicity. Although it has been previously shown that chronic application of lactate (6 h) causes increased gene transcription and protein expression, we observe clustering of TREK1 channels that is dependent on time of exposure (3-6 h) and concentration of lactate (15-30 mM). Using immunofluorescence techniques and image analysis, we show that the clustering of TREK1 channels is dependent on the actin cytoskeletal network of the astrocytes. Clustering of TREK1 channels can augment astrocytic functions during pathophysiological conditions and have significant implications in lactate mediated neuroprotection.
Collapse
Affiliation(s)
- Swagata Ghatak
- School of Biological Sciences, National Institute of Science Education and Research (NISER)-Bhubaneswar, an OCC of Homi Bhabha National Institute, Jatani, Odisha 752050, India; Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka 560012, India.
| | - Sujit Kumar Sikdar
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka 560012, India.
| |
Collapse
|
7
|
Perosa V, Zanon Zotin MC, Schoemaker D, Sveikata L, Etherton MR, Charidimou A, Greenberg SM, Viswanathan A. Association Between Hippocampal Volumes and Cognition in Cerebral Amyloid Angiopathy. Neurology 2024; 102:e207854. [PMID: 38165326 PMCID: PMC10870737 DOI: 10.1212/wnl.0000000000207854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Accumulating evidence suggests that gray matter atrophy, often considered a marker of Alzheimer disease (AD), can also result from cerebral small vessel disease (CSVD). Cerebral amyloid angiopathy (CAA) is a form of sporadic CSVD, diagnosed through neuroimaging criteria, that often co-occurs with AD pathology and leads to cognitive impairment. We sought to identify the role of hippocampal integrity in the development of cognitive impairment in a cohort of patients with possible and probable CAA. METHODS Patients were recruited from an ongoing CAA study at Massachusetts General Hospital. Composite scores defined performance in the cognitive domains of memory, language, executive function, and processing speed. Hippocampal subfields' volumes were measured from 3T MRI, using an automated method, and multivariate linear regression models were used to estimate their association with each cognitive domain and relationship to CAA-related neuroimaging markers. RESULTS One hundred twenty patients, 36 with possible (age mean [range]: 75.6 [65.6-88.9]), 67 with probable CAA (75.9 [59.0-94.0]), and 17 controls without cognitive impairment and CSVD (72.4 [62.5-82.7]; 76.4% female patients), were included in this study. We found a positive association between all investigated hippocampal subfields and memory and language, whereas specific subfields accounted for executive function (CA4 [Estimate = 5.43; 95% CI 1.26-9.61; p = 0.020], subiculum [Estimate = 2.85; 95% CI 0.67-5.02; p = 0.022]), and processing speed (subiculum [Estimate = 1.99; 95% CI 0.13-3.85; p = 0.036]). These findings were independent of other CAA-related markers, which did not have an influence on cognition in this cohort. Peak width of skeletonized mean diffusivity (PSMD), a measure of white matter integrity, was negatively associated with hippocampal subfields' volumes (CA3 [Estimate = -0.012; 95% CI -0.020 to -0.004; p = 0.034], CA4 [Estimate = -0.010; 95% CI -0.020 to -0.0007; p = 0.037], subiculum [Estimate = -0.019; 95% CI -0.042 to -0.0001; p = 0.003]). DISCUSSION These results suggest that hippocampal integrity is an independent contributor to cognitive impairment in patients with CAA and that it might be related to loss of integrity in the white matter. Further studies exploring potential causes and directionality of the relationship between white matter and hippocampal integrity may be warranted.
Collapse
Affiliation(s)
- Valentina Perosa
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Maria Clara Zanon Zotin
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Dorothee Schoemaker
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lukas Sveikata
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mark R Etherton
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Andreas Charidimou
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Steven M Greenberg
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Anand Viswanathan
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
8
|
Sadeghzadeh J, Hosseini L, Mobed A, Zangbar HS, Jafarzadeh J, Pasban J, Shahabi P. The Impact of Cerebral Ischemia on Antioxidant Enzymes Activity and Neuronal Damage in the Hippocampus. Cell Mol Neurobiol 2023; 43:3915-3928. [PMID: 37740074 DOI: 10.1007/s10571-023-01413-w] [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: 02/27/2023] [Accepted: 09/09/2023] [Indexed: 09/24/2023]
Abstract
Cerebral ischemia and subsequent reperfusion, leading to reduced blood supply to specific brain areas, remain significant contributors to neurological damage, disability, and mortality. Among the vulnerable regions, the subcortical areas, including the hippocampus, are particularly susceptible to ischemia-induced injuries, with the extent of damage influenced by the different stages of ischemia. Neural tissue undergoes various changes and damage due to intricate biochemical reactions involving free radicals, oxidative stress, inflammatory responses, and glutamate toxicity. The consequences of these processes can result in irreversible harm. Notably, free radicals play a pivotal role in the neuropathological mechanisms following ischemia, contributing to oxidative stress. Therefore, the function of antioxidant enzymes after ischemia becomes crucial in preventing hippocampal damage caused by oxidative stress. This study explores hippocampal neuronal damage and enzymatic antioxidant activity during ischemia and reperfusion's early and late stages.
Collapse
Affiliation(s)
- Jafar Sadeghzadeh
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Leila Hosseini
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Ahmad Mobed
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Jaber Jafarzadeh
- Department of Community Nutrition Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Jamshid Pasban
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Parviz Shahabi
- Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.
| |
Collapse
|
9
|
Noorgaldi S, Sarkala HB, Enayati A, Khori V, Zengin G, Jahanshahi M. Neuroprotective effect of Potentilla reptans L. root in the rat brain global ischemia/reperfusion model. Arch Pharm (Weinheim) 2023; 356:e2300363. [PMID: 37642540 DOI: 10.1002/ardp.202300363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
Stroke is the most common cause of death among neurological diseases. The protective effects of Potentilla reptans L. include antioxidative, anti-inflammatory, and antiapoptotic effects. In this study, the brain protection and beta-amyloid effects of P. reptans root extract were investigated in the rat brain ischemia/reperfusion (IR) model. Forty male Wistar rats were randomly divided into five groups (n = 8), including IR, sham, and three groups receiving P. reptans with concentrations of 0.025, 0.05, and 0.1 (g/kg/b.w.), which were injected daily for 7 days. For the IR model, the common carotid artery was occluded bilaterally for 8 min. All injections were intraperitoneal (IP). The shuttle box test was used to measure passive avoidance memory. Then the brain tissue was extracted for the histological examination of neuron counts and β-amyloid plaques using a morphometric technique, and finally, Statistical Package for the Social Sciences software was used for statistical analysis of the data. Pretreatment with P. reptans improved memory impairment. Also, by examining the tissues of the CA1, CA3, and dentate gyrus areas of the hippocampus, it was observed that the number of plaques in the groups receiving P. reptans extract was reduced compared to the IR group, especially at the concentration of 0.05 g/kg/b.w. Also, P. reptans improved the number of neurons at all concentrations, in which the concentration of 0.05 g/kg/b.w. showed more effective therapeutic results. Taken together, we found that P. reptans root extract has beneficial effects on memory impairment, neuronal loss, and β-amyloid accumulation.
Collapse
Affiliation(s)
- Soraya Noorgaldi
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Anatomy, Faculty of Medicine, Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hamzeh Badeli Sarkala
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ayesheh Enayati
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Gökhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Mehrdad Jahanshahi
- Department of Anatomy, Faculty of Medicine, Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
10
|
Zhang X, Yang SB, Cheng L, Ho K, Kim MS. Botanical Mixture Containing Nitric Oxide Metabolite Enhances Neural Plasticity to Improve Cognitive Impairment in a Vascular Dementia Rat Model. Nutrients 2023; 15:4381. [PMID: 37892455 PMCID: PMC10609983 DOI: 10.3390/nu15204381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/12/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Vascular dementia (VD), caused by impaired cerebral blood flow, is the most common form of dementia after Alzheimer's disease (AD) in the elderly and is characterized by severe neuronal damage and cognitive decline. Nitric oxide (NO) is an important determinant of vascular homeostasis, and its deficiency is associated with the progression of VD. In this study, we investigated the role of nitrite ion, a NO metabolite in a botanical mixture (BM) of fermented garlic, fermented Scutellaria baicalensis, and Rhodiola rosea on neuron loss and cognitive impairment using a VD rat model. The BM containing the NO metabolite alleviated cognitive deficits and enhanced neural plasticity, as reflected by an increase in long-term potentiation. The BM also alleviated neuron apoptosis, decreased GFAP expression, and oxidative stress, and increased parvalbumin and brain-derived neurotrophic factor (BDNF) levels. These results indicate that BM exerts neuroprotective effects and alleviates cognitive dysfunction while enhancing neuroplasticity, and thus has therapeutic potential against VD.
Collapse
Affiliation(s)
- Xiaorong Zhang
- Department of Pathology, Affiliated Hospital of Jiujiang University, Jiujiang 332000, China
- Center for Cognitive Science and Transdisciplinary Studies, Jiujiang University, Jiujiang 332000, China
- Center for Nitric Oxide Metabolite, Wonkwang University, Iksan 54538, Republic of Korea
| | - Seung-Bum Yang
- Department of Medical Non-Commissioned Officer, Wonkwang Health Science University, Iksan 54538, Republic of Korea
| | - Lin Cheng
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang 332000, China
| | - Koo Ho
- Center for Nitric Oxide Metabolite, Wonkwang University, Iksan 54538, Republic of Korea
| | - Min-Sun Kim
- Center for Nitric Oxide Metabolite, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
11
|
Escobar I, Xu J, Jackson CW, Stegelmann SD, Fagerli EA, Dave KR, Perez-Pinzon MA. Resveratrol Preconditioning Protects Against Ischemia-Induced Synaptic Dysfunction and Cofilin Hyperactivation in the Mouse Hippocampal Slice. Neurotherapeutics 2023; 20:1177-1197. [PMID: 37208551 PMCID: PMC10457274 DOI: 10.1007/s13311-023-01386-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/21/2023] Open
Abstract
Perturbations in synaptic function are major determinants of several neurological diseases and have been associated with cognitive impairments after cerebral ischemia (CI). Although the mechanisms underlying CI-induced synaptic dysfunction have not been well defined, evidence suggests that early hyperactivation of the actin-binding protein, cofilin, plays a role. Given that synaptic impairments manifest shortly after CI, prophylactic strategies may offer a better approach to prevent/mitigate synaptic damage following an ischemic event. Our laboratory has previously demonstrated that resveratrol preconditioning (RPC) promotes cerebral ischemic tolerance, with many groups highlighting beneficial effects of resveratrol treatment on synaptic and cognitive function in other neurological conditions. Herein, we hypothesized that RPC would mitigate hippocampal synaptic dysfunction and pathological cofilin hyperactivation in an ex vivo model of ischemia. Various electrophysiological parameters and synaptic-related protein expression changes were measured under normal and ischemic conditions utilizing acute hippocampal slices derived from adult male mice treated with resveratrol (10 mg/kg) or vehicle 48 h prior. Remarkably, RPC significantly increased the latency to anoxic depolarization, decreased cytosolic calcium accumulation, prevented aberrant increases in synaptic transmission, and rescued deficits in long-term potentiation following ischemia. Additionally, RPC upregulated the expression of the activity-regulated cytoskeleton associated protein, Arc, which was partially required for RPC-mediated attenuation of cofilin hyperactivation. Taken together, these findings support a role for RPC in mitigating CI-induced excitotoxicity, synaptic dysfunction, and pathological over-activation of cofilin. Our study provides further insight into mechanisms underlying RPC-mediated neuroprotection against CI and implicates RPC as a promising strategy to preserve synaptic function after ischemia.
Collapse
Affiliation(s)
- Iris Escobar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Jing Xu
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Charles W Jackson
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Samuel D Stegelmann
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Eric A Fagerli
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, PO Box 016960, Miami, FL, 33101, USA.
| |
Collapse
|
12
|
Srakočić S, Gorup D, Kutlić D, Petrović A, Tarabykin V, Gajović S. Reactivation of corticogenesis-related transcriptional factors BCL11B and SATB2 after ischemic lesion of the adult mouse brain. Sci Rep 2023; 13:8539. [PMID: 37237015 DOI: 10.1038/s41598-023-35515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The aim of this study was to characterize expression of corticogenesis-related transcription factors BCL11B and SATB2 after brain ischemic lesion in the adult mice, and to analyze their correlation to the subsequent brain recovery. Ischemic brain lesion was induced by transient middle cerebral artery occlusion followed by reperfusion, and the animals with ischemic lesion were compared to the sham controls. Progression of the brain damage and subsequent recovery was longitudinally monitored structurally, by magnetic resonance imaging, and functionally, by neurological deficit assessment. Seven days after the ischemic injury the brains were isolated and analyzed by immunohistochemistry. The results showed higher expression in the brain of both, BCL11B and SATB2 in the animals with ischemic lesion compared to the sham controls. The co-expression of both markers, BCL11B and SATB2, increased in the ischemic brains, as well as the co-expression of BCL11B with the beneficial transcriptional factor ATF3 but not its co-expression with detrimental HDAC2. BCL11B was mainly implicated in the ipsilateral and SATB2 in the contralateral brain hemisphere, and their level in these regions correlated with the functional recovery rate. The results indicate that the reactivation of corticogenesis-related transcription factors BCL11B and SATB2 is beneficial after brain ischemic lesion.
Collapse
Affiliation(s)
- Sanja Srakočić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Dunja Gorup
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
- Universität Zürich, Universitätspital Zürich, Zürich, Switzerland
| | - Dominik Kutlić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Ante Petrović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin, Berlin, Germany
- Institute of Neuroscience, University of Nizhny Novgorod, Pr. Gagarina 24, Nizhny Novgorod, Russia
| | - Srećko Gajović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia.
| |
Collapse
|
13
|
Ferrier FJ, Saul I, Khoury N, Ruiz AJ, Lao EJP, Escobar I, Dave KR, Young JI, Perez-Pinzon MA. Post cardiac arrest physical exercise mitigates cell death in the septal and thalamic nuclei and ameliorates contextual fear conditioning deficits in rats. J Cereb Blood Flow Metab 2023; 43:446-459. [PMID: 36369732 PMCID: PMC9941858 DOI: 10.1177/0271678x221137539] [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: 04/28/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/14/2022]
Abstract
A major concern for cardiac arrest (CA) survivors is the manifestation of long-term cognitive impairments. Physical exercise (PE) is a well-established approach to improve cognitive functions under certain pathological conditions. We previously showed that PE post-CA mitigates cognitive deficits, but the underlying mechanisms remain unknown. To define neuroprotective mechanisms, we analyzed whether PE post-CA protects neurons involved in memory. We first performed a contextual fear conditioning (CFC) test to confirm that PE post-CA preserves memory in rats. We then conducted a cell-count analysis and determined the number of live cells in the hippocampus, and septal and thalamic nuclei, all areas involved in cognitive functions. Lastly, we performed RNA-seq to determine PE post-CA effect on gene expression. Following CA, exercised rats had preserved CFC memory than sham PE animals. Despite this outcome, PE post-CA did not protect hippocampal cells from dying. However, PE ameliorated cell death in septal and thalamic nuclei compared to sham PE animals, suggesting that these nuclei are crucial in mitigating cognitive decline post-CA. Interestingly, PE affected regulation of genes related to neuroinflammation, plasticity, and cell death. These findings reveal potential mechanisms whereby PE post-CA preserves cognitive functions by protecting septal and thalamic cells via gene regulation.
Collapse
Affiliation(s)
- Fernando J Ferrier
- Peritz Scheinberg Cerebral Vascular Disease Research
Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami,
FL, USA
- Neuroscience Program, University of Miami Leonard M. Miller
School of Medicine, Miami FL
| | - Isabel Saul
- Peritz Scheinberg Cerebral Vascular Disease Research
Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami,
FL, USA
- Department of Neurology, University of Miami Leonard M. Miller
School of Medicine, Miami, FL, USA
| | - Nathalie Khoury
- Peritz Scheinberg Cerebral Vascular Disease Research
Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami,
FL, USA
- Neuroscience Program, University of Miami Leonard M. Miller
School of Medicine, Miami FL
| | - Alexander J Ruiz
- Peritz Scheinberg Cerebral Vascular Disease Research
Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami,
FL, USA
| | - Efrain J Perez Lao
- Peritz Scheinberg Cerebral Vascular Disease Research
Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami,
FL, USA
- Neuroscience Program, University of Miami Leonard M. Miller
School of Medicine, Miami FL
- Hussman Institute for Human Genetics, University of Miami
Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Iris Escobar
- Peritz Scheinberg Cerebral Vascular Disease Research
Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami,
FL, USA
- Neuroscience Program, University of Miami Leonard M. Miller
School of Medicine, Miami FL
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research
Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami,
FL, USA
- Neuroscience Program, University of Miami Leonard M. Miller
School of Medicine, Miami FL
- Department of Neurology, University of Miami Leonard M. Miller
School of Medicine, Miami, FL, USA
| | - Juan I Young
- Hussman Institute for Human Genetics, University of Miami
Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research
Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami,
FL, USA
- Neuroscience Program, University of Miami Leonard M. Miller
School of Medicine, Miami FL
- Department of Neurology, University of Miami Leonard M. Miller
School of Medicine, Miami, FL, USA
| |
Collapse
|
14
|
Johnson AC. Hippocampal Vascular Supply and Its Role in Vascular Cognitive Impairment. Stroke 2023; 54:673-685. [PMID: 36848422 PMCID: PMC9991081 DOI: 10.1161/strokeaha.122.038263] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/22/2022] [Indexed: 03/01/2023]
Abstract
The incidence of age-related dementia is increasing as the world population ages and due to lack of effective treatments for dementia. Vascular contributions to cognitive impairment and dementia are increasing as the prevalence of pathologies associated with cerebrovascular disease rise, including chronic hypertension, diabetes, and ischemic stroke. The hippocampus is a bilateral deep brain structure that is central to learning, memory, and cognitive function and highly susceptible to hypoxic/ischemic injury. Compared with cortical brain regions such as the somatosensory cortex, less is known about the function of the hippocampal vasculature that is critical in maintaining neurocognitive health. This review focuses on the hippocampal vascular supply, presenting what is known about hippocampal hemodynamics and blood-brain barrier function during health and disease, and discusses evidence that supports its contribution to vascular cognitive impairment and dementia. Understanding vascular-mediated hippocampal injury that contributes to memory dysfunction during healthy aging and cerebrovascular disease is essential to develop effective treatments to slow cognitive decline. The hippocampus and its vasculature may represent one such therapeutic target to mitigate the dementia epidemic.
Collapse
Affiliation(s)
- Abbie C Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington
| |
Collapse
|
15
|
Zhang R, Chen M, Deng Z, Kong L, Shen B, Zhang L. Delta Opioid Peptide Targets Brain Microvascular Endothelial Cells Reducing Apoptosis to Relieve Hypoxia-Ischemic/Reperfusion Injury. Pharmaceutics 2022; 15:pharmaceutics15010046. [PMID: 36678674 PMCID: PMC9861451 DOI: 10.3390/pharmaceutics15010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is one of the leading causes of death. (D-ala2, D-leu5) enkephalin (DADLE) is a synthetic peptide and highly selective delta opioid receptor (δOR) agonist that has exhibited protective properties in ischemia. However, the specific target and mechanism are still unclear. The present study explores the expression of δOR on brain microvascular endothelial cells (BMECs) and whether DADLE could relieve I/R-induced injury by reducing apoptosis. A lateral ventricular injection of DADLE for pretreatment, the neurofunctional behavior score, and TTC staining, were used to evaluate the protective effect of DADLE. Immunofluorescence technology was used to label different types of cells with apoptosis-positive signals to test co-localization status. Primary cultured BMECs were separated and treated with DADLE, accompanied by OGD/R. The CCK-8 test was conducted to evaluate cell viability and TdT-mediated dUTP Nick-end Labelling (TUNEL) staining to test apoptosis levels. The levels of apoptosis-related proteins were analyzed by Western blotting. The co-localization results showed that BMECs, but not astrocytes, microglia, or neurons, presented mostly TUNEL-positive signals, especially in the Dentate gyrus (DG) area of the hippocampus. Either activation of δORs on rats' brains or primary BMECs mainly reduce cellular apoptosis and relieve the injury. Interference with the expression δOR could block this effect. DADLE also significantly increased levels of Bcl-2 and reduced levels of Bax. δOR's expressions can be detected on the BMECs, but not on the HEK293 cells, by Western blotting and IFC. Therefore, DADLE exerts a cytoprotective effect, primarily under hypoxia-ischemic injury/reperfusion conditions, by targeting BMECs to inhibit apoptosis.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Meixuan Chen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhongfang Deng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lingchao Kong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Bing Shen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lesha Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Correspondence:
| |
Collapse
|
16
|
Baranovicova E, Kalenska D, Kovalska M, Lehotsky J. Hippocampal metabolic recovery as a manifestation of the protective effect of ischemic preconditioning in rats. Neurochem Int 2022; 160:105419. [PMID: 36113578 DOI: 10.1016/j.neuint.2022.105419] [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: 02/21/2022] [Revised: 08/25/2022] [Accepted: 09/04/2022] [Indexed: 10/31/2022]
Abstract
The ever-present risk of brain ischemic events in humans and its full prevention make the detailed studies of an organism's response to ischemia at different levels essential to understanding the mechanism of the injury as well as protection. We used the four-vessel occlusion as an animal model of forebrain ischemia to investigate its impact on the metabolic alterations in both the hippocampus and the blood plasma to see changes on the systemic level. By inducing sublethal ischemic stimuli, we focused on the endogenous phenomena known as ischemic tolerance. NMR spectroscopy was used to analyze relative metabolite levels in tissue extracts from rats' hippocampus and blood plasma in three various ischemic/reperfusion times: 3 h, 24 h, and 72 h. Hippocampal tissues were characterized by postischemically decreased glutamate and GABA (4-aminobutyrate) tissue content balanced with increased glutamine level, with most pronounced changes at 3 h reperfusion time. Glutamate (as well as glutamine) levels recovered towards the control levels on the third day, as if the glutamate re-synthesis would be firstly preferred before GABA. These results are indicating the higher feasibility of re-establishing of glutamatergic transmission three days after an ischemic event, in contrast to GABA-ergic. Tissue levels of N-acetylaspartate (NAA), as well as choline, were decreased without the tendency to recover three days after the ischemic event. Metabolomic analysis of blood plasma revealed that ischemically preconditioned rats, contrary to the non-preconditioned animals, did not show hyperglycemic conditions. Ischemically induced semi-ketotic state, manifested in increased plasma ketone bodies 3-hydroxybutyrate and acetoacetate, seems to be programmed to support the brain tissue revitalization after the ischemic event. These and other metabolites changes found in blood plasma as well as in the hippocampus were observed to a lower extent or recovered faster in preconditioned animals. Some metabolomic changes in hippocampal tissue extract were so strong that even single metabolites were able to differentiate between ischemic, ischemically preconditioned, and control brain tissues.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| |
Collapse
|
17
|
Johnson AC, Uhlig F, Einwag Z, Cataldo N, Erdos B. The neuroendocrine stress response impairs hippocampal vascular function and memory in male and female rats. Neurobiol Dis 2022; 168:105717. [PMID: 35385769 PMCID: PMC9018625 DOI: 10.1016/j.nbd.2022.105717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 11/24/2022] Open
Abstract
Chronic psychological stress affects brain regions involved in memory such as the hippocampus and accelerates age-related cognitive decline, including in Alzheimer's disease and vascular dementia. However, little is known about how chronic stress impacts hippocampal vascular function that is critically involved in maintaining neurocognitive health that could contribute to stress-related memory dysfunction. Here, we used a novel experimental rat model that mimics the neuroendocrine and cardiovascular aspects of chronic stress to determine how the neuroendocrine components of the stress response affect hippocampal function. We studied both male and female rats to determine potential sex differences in the susceptibility of the hippocampus and its vasculature to neuroendocrine stress-induced dysfunction. We show that activation of neuroendocrine stress pathways impaired the vasoreactivity of hippocampal arterioles to mediators involved in coupling neuronal activity with local blood flow that was associated with impaired memory function. Interestingly, we found more hippocampal arteriolar dysfunction and scarcer hippocampal microvasculature in male compared to female rats that was associated with greater memory impairment, suggesting the male sex may be at increased risk of neuroendocrine-derived hippocampal dysfunction during chronic stress. Overall, this study revealed the therapeutic potential of targeting hippocampal arterioles to prevent or slow memory decline in the setting of prolonged and/or unavoidable stress.
Collapse
Affiliation(s)
- Abbie C Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA.
| | - Friederike Uhlig
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Zachary Einwag
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Noelle Cataldo
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| | - Benedek Erdos
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, 05405, USA
| |
Collapse
|
18
|
Magierowska K, Korbut E, Wójcik-Grzybek D, Bakalarz D, Sliwowski Z, Cieszkowski J, Szetela M, Torregrossa R, Whiteman M, Magierowski M. Mitochondria-targeted hydrogen sulfide donors versus acute oxidative gastric mucosal injury. J Control Release 2022; 348:321-334. [PMID: 35654168 DOI: 10.1016/j.jconrel.2022.05.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/14/2022]
Abstract
Hydrogen sulfide (H2S) as a gaseous molecule prevents gastrointestinal (GI)-tract against various injuries. This study aimed to evaluate for the first time the detailed molecular mechanism of mitochondria-targeting H2S-prodrugs, AP39 and RT01 in gastroprotection against ischemia/reperfusion (I/R)-induced lesions. Wistar rats exposed to I/R were pretreated i.g. with vehicle, AP39 (0.004-2 mg/kg), RT01 (0.1 mg/kg), or with AP219 (0.1 mg/kg) as structural control without ability to release H2S. AP39 was also administered with mTOR1 inhibitor, rapamycin (1 mg/kg i.g.). Gastric damage area was assessed micro-/macroscopically, gastric blood flow (GBF) by laser flowmetry, mRNA level of HIF-1α, GPx, SOD1, SOD2, annexin-A1, SOCS3, IL-1RA, IL-1β, IL-1R1, IL-1R2, TNFR2, iNOS by real-time PCR. Gastric mucosal and/or serum content of IL-1β, IL-4, IL-5, IL-10, G-CSF, M-CSF, VEGFA, GRO, RANTES, MIP-1α, MCP1, TNF-α, TIMP1, FABP3, GST-α, STAT3/5 and phosphorylation of mTOR, NF-κB, ERK, Akt was evaluated by microbeads-fluorescent assay. Mitochondrial complexes activities were measured biochemically. RNA damage was assessed as 8-OHG by ELISA. AP39 and RT01 reduced micro-/macroscopic gastric I/R-injury increasing GBF. AP39-gastroprotection was accompanied by maintained activity of mitochondrial complexes, prevented RNA oxidation and enhanced mRNA/protein expression of SOCS3, IL-1RA, annexin-A1, GST-α, HIF-1α. Rapamycin reversed AP-39-gastroprotection. AP39-gastroprotection was followed by decreased NF-κB, ERK, IL-1β and enhanced Akt and mTOR proteins phosphorylation. AP39-prevented gastric mucosal damage caused by I/R-injury, partly by mitochondrial complex activity maintenance. AP39-mediated attenuation of gastric mucosal oxidation, hypoxia and inflammation involved mTOR1 and Akt pathways activity and modulation of HIF-1α, GST-α, SOCS3, IL1RA and TIMP1 molecular interplay.
Collapse
Affiliation(s)
| | - Edyta Korbut
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | | | - Dominik Bakalarz
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland; Department of Forensic Toxicology, Institute of Forensic Research, Cracow, Poland
| | - Zbigniew Sliwowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Jakub Cieszkowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Małgorzata Szetela
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | | | | | - Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland.
| |
Collapse
|
19
|
Liao S, Luo Y, Chunchai T, Singhanat K, Arunsak B, Benjanuwattra J, Apaijai N, Chattipakorn N, Chattipakorn SC. An apoptosis inhibitor suppresses microglial and astrocytic activation after cardiac ischemia/reperfusion injury. Inflamm Res 2022; 71:861-872. [PMID: 35655102 DOI: 10.1007/s00011-022-01590-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 05/06/2022] [Accepted: 05/16/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Microglial hyperactivation and apoptosis were observed following myocardial infarction and ischemia reperfusion (I/R) injury. This study aimed to test the hypothesis that the apoptosis inhibitor, Z-VAD, attenuates microglial and astrocytic hyperactivation and brain inflammation in rats with cardiac I/R injury. MATERIALS AND METHODS Rats were subjected to either sham or cardiac I/R operation (30 min-ischemia followed by 120-min reperfusion), rats in the cardiac I/R group were given either normal saline solution or Z-VAD at 3.3 mg/kg via intravenous injection 15 min prior to cardiac ischemia. Left ventricular ejection fraction (% LVEF) was determined during the cardiac I/R protocol. The brain tissues were removed and used to determine brain apoptosis, brain inflammation, microglial and astrocyte morphology. RESULTS Cardiac dysfunction was observed in rats with cardiac I/R injury as indicated by decreased %LVEF. In the brain, we found brain apoptosis, brain inflammation, microglia hyperactivation, and reactive astrogliosis occurred following cardiac I/R injury. Pretreatment with Z-VAD effectively increased %LVEF, reduced brain apoptosis, attenuated brain inflammation by decreasing IL-1β mRNA levels, suppressed microglial and astrocytic hyperactivation and proliferation after cardiac I/R injury. CONCLUSION Z-VAD exerts neuroprotective effects against cardiac I/R injury not only targeting apoptosis but also microglial and astrocyte activation.
Collapse
Affiliation(s)
- Suchan Liao
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Ying Luo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Titikorn Chunchai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Kodchanan Singhanat
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Busarin Arunsak
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Juthipong Benjanuwattra
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
20
|
Abstract
BACKGROUND Blood pressure variability (BPV) has been linked with cognitive impairment and dementia. However, the pathophysiological mechanisms by which BPV affects cognition are unclear. This systematic review aims to assess the links between different BPV measures and white and grey matter structures. METHODS AND RESULTS The following databases were searched from inception through to January 2021; EMBASE, MEDLINE, EMCARE and SCOPUS. Studies that reported on the relationship between within-individual BPV (short, medium or long-term variability) or a circadian blood pressure (BP) measurement and MRI assessed brain structures were included. Overall, 20 studies met the criteria and were included, of which 11 studies looked at short-term BPV, eight articles investigated visit-to-visit BPV and one study looked at a compositional BPV measurement. Due to heterogeneity in study samples, meta-analysis was not possible. Across the included studies, associations between MRI indices and BP dipping patterns were mixed; higher long-term BPV and higher sleep systolic BPV was found to be associated with lower whole brain volume and hippocampal volume. CONCLUSION Increased BPV, in particular systolic long-term and systolic night-time BPV, appears to be associated with lower brain volume and hippocampal volume. This highlights the adverse effect that increased BPV has upon the brain, potentially contributing to cognitive decline, including dementia, in late-life.
Collapse
|
21
|
Zavaliangos‐Petropulu A, Lo B, Donnelly MR, Schweighofer N, Lohse K, Jahanshad N, Barisano G, Banaj N, Borich MR, Boyd LA, Buetefisch CM, Byblow WD, Cassidy JM, Charalambous CC, Conforto AB, DiCarlo JA, Dula AN, Egorova‐Brumley N, Etherton MR, Feng W, Fercho KA, Geranmayeh F, Hanlon CA, Hayward KS, Hordacre B, Kautz SA, Khlif MS, Kim H, Kuceyeski A, Lin DJ, Liu J, Lotze M, MacIntosh BJ, Margetis JL, Mohamed FB, Piras F, Ramos‐Murguialday A, Revill KP, Roberts PS, Robertson AD, Schambra HM, Seo NJ, Shiroishi MS, Stinear CM, Soekadar SR, Spalletta G, Taga M, Tang WK, Thielman GT, Vecchio D, Ward NS, Westlye LT, Werden E, Winstein C, Wittenberg GF, Wolf SL, Wong KA, Yu C, Brodtmann A, Cramer SC, Thompson PM, Liew S. Chronic Stroke Sensorimotor Impairment Is Related to Smaller Hippocampal Volumes: An ENIGMA Analysis. J Am Heart Assoc 2022; 11:e025109. [PMID: 35574963 PMCID: PMC9238563 DOI: 10.1161/jaha.121.025109] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
Abstract
Background Persistent sensorimotor impairments after stroke can negatively impact quality of life. The hippocampus is vulnerable to poststroke secondary degeneration and is involved in sensorimotor behavior but has not been widely studied within the context of poststroke upper-limb sensorimotor impairment. We investigated associations between non-lesioned hippocampal volume and upper limb sensorimotor impairment in people with chronic stroke, hypothesizing that smaller ipsilesional hippocampal volumes would be associated with greater sensorimotor impairment. Methods and Results Cross-sectional T1-weighted magnetic resonance images of the brain were pooled from 357 participants with chronic stroke from 18 research cohorts of the ENIGMA (Enhancing NeuoImaging Genetics through Meta-Analysis) Stroke Recovery Working Group. Sensorimotor impairment was estimated from the FMA-UE (Fugl-Meyer Assessment of Upper Extremity). Robust mixed-effects linear models were used to test associations between poststroke sensorimotor impairment and hippocampal volumes (ipsilesional and contralesional separately; Bonferroni-corrected, P<0.025), controlling for age, sex, lesion volume, and lesioned hemisphere. In exploratory analyses, we tested for a sensorimotor impairment and sex interaction and relationships between lesion volume, sensorimotor damage, and hippocampal volume. Greater sensorimotor impairment was significantly associated with ipsilesional (P=0.005; β=0.16) but not contralesional (P=0.96; β=0.003) hippocampal volume, independent of lesion volume and other covariates (P=0.001; β=0.26). Women showed progressively worsening sensorimotor impairment with smaller ipsilesional (P=0.008; β=-0.26) and contralesional (P=0.006; β=-0.27) hippocampal volumes compared with men. Hippocampal volume was associated with lesion size (P<0.001; β=-0.21) and extent of sensorimotor damage (P=0.003; β=-0.15). Conclusions The present study identifies novel associations between chronic poststroke sensorimotor impairment and ipsilesional hippocampal volume that are not caused by lesion size and may be stronger in women.
Collapse
Affiliation(s)
- Artemis Zavaliangos‐Petropulu
- Mark and Mary Stevens Neuroimaging and Informatics InstituteKeck School of Medicine, University of Southern CaliforniaLos AngelesCA
- Neuroscience Graduate ProgramUniversity of Southern CaliforniaLos AngelesCA
| | - Bethany Lo
- Chan Division of Occupational Science and Occupational TherapyUniversity of Southern CaliforniaLos AngelesCA
| | - Miranda R. Donnelly
- Chan Division of Occupational Science and Occupational TherapyUniversity of Southern CaliforniaLos AngelesCA
| | - Nicolas Schweighofer
- Biokinesiology and Physical TherapyUniversity of Southern CaliforniaLos AngelesCA
| | - Keith Lohse
- Physical Therapy and NeurologyWashington University School of Medicine in Saint LouisMO
| | - Neda Jahanshad
- Mark and Mary Stevens Neuroimaging and Informatics InstituteKeck School of Medicine, University of Southern CaliforniaLos AngelesCA
| | - Giuseppe Barisano
- Mark and Mary Stevens Neuroimaging and Informatics InstituteKeck School of Medicine, University of Southern CaliforniaLos AngelesCA
- Neuroscience Graduate ProgramUniversity of Southern CaliforniaLos AngelesCA
| | - Nerisa Banaj
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Michael R. Borich
- Division of Physical TherapyDepartment of Rehabilitation MedicineEmory University School of MedicineAtlantaGA
| | - Lara A. Boyd
- Department of Physical TherapyUniversity of British ColumbiaVancouverCanada
| | | | - Winston D. Byblow
- Department of Exercise Sciences, and Centre for Brain ResearchUniversity of AucklandNew Zealand
| | - Jessica M. Cassidy
- Department of Allied Health SciencesUniversity of North Carolina at Chapel HillNC
| | - Charalambos C. Charalambous
- Department of Basic and Clinical SciencesUniversity of Nicosia Medical SchoolNicosiaCyprus
- Center for Neuroscience and Integrative Brain Research (CENIBRE)NicosiaCyprus
| | - Adriana B. Conforto
- Hospital das ClínicasSão Paulo UniversitySão PauloBrazil
- Hospital Israelita Albert EinsteinSão PauloBrazil
| | - Julie A. DiCarlo
- Center for Neurotechnology and Neurorecovery (CNTR)Massachusetts General HospitalBostonMA
| | - Adrienne N. Dula
- Department of NeurologyDell Medical SchoolUniversity of Texas at AustinTX
| | | | - Mark R. Etherton
- Department of NeurologyJ. Philip Kistler Stroke Research CenterMassachusetts General HospitalBostonMA
| | - Wuwei Feng
- Department of NeurologyDuke University School of MedicineDurhamNC
| | - Kelene A. Fercho
- Basic Biomedical SciencesUniversity of South DakotaVermillionSD
- Federal Aviation AdministrationCivil Aerospace Medical InstituteOklahoma CityOK
| | | | | | - Kathryn S. Hayward
- Departments of Physiotherapy and Medicine, University of MelbourneHeidelbergVictoriaAustralia
- The Florey Institute of Neuroscience and Mental HealthHeidelbergVictoriaAustralia
| | - Brenton Hordacre
- Innovation, Implementation and Clinical Translation (IIMPACT) in HealthAllied Health and Human PerformanceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Steven A. Kautz
- Ralph H Johnson Veterans Affairs Medical CenterCharlestonSC
- Department of Health Sciences & ResearchMedical University of South CarolinaCharlestonSC
| | - Mohamed Salah Khlif
- The Florey Institute of Neuroscience and Mental HealthHeidelbergVictoriaAustralia
| | - Hosung Kim
- Mark and Mary Stevens Neuroimaging and Informatics InstituteKeck School of Medicine, University of Southern CaliforniaLos AngelesCA
| | - Amy Kuceyeski
- Department of RadiologyWeill Cornell MedicineNew YorkNY
| | - David J. Lin
- Center for Neurotechnology and Neurorecovery (CNTR)Massachusetts General HospitalBostonMA
| | - Jingchun Liu
- Department of RadiologyTianjin Medical University General HospitalTianjinChina
| | - Martin Lotze
- Functional ImagingInstitute for Diagnostic Radiology and NeuroradiologyUniversity Medicine GreifswaldGermany
| | - Bradley J. MacIntosh
- Hurvitz Brain Sciences ProgramSunnybrook Research InstituteTorontoCanada
- Department of Medical BiophysicsUniversity of TorontoOntarioCanada
| | - John L. Margetis
- Chan Division of Occupational Science and Occupational TherapyUniversity of Southern CaliforniaLos AngelesCA
| | - Feroze B. Mohamed
- Department of RadiologyJefferson Integrated MR CenterThomas Jefferson UniversityPhiladelphiaPA
| | - Fabrizio Piras
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Ander Ramos‐Murguialday
- Institute of Medical Psychology and Behavioral NeurobiologyUniversity of TübingenGermany
- Health DivisionTECNALIASan SebastianSpain
| | | | - Pamela S. Roberts
- Chan Division of Occupational Science and Occupational TherapyUniversity of Southern CaliforniaLos AngelesCA
- Department of Physical Medicine and RehabilitationCedars‐SinaiLos AngelesCA
| | - Andrew D. Robertson
- Department of Kinesiology and Health SciencesUniversity of WaterlooOntarioCanada
| | - Heidi M. Schambra
- Departments of Neurology & Rehabilitation MedicineNYU LangoneNew YorkNY
| | - Na Jin Seo
- Ralph H Johnson Veterans Affairs Medical CenterCharlestonSC
- Department of Rehabilitation SciencesDepartment of Health Science and ResearchMedical University of South CarolinaCharlestonSC
| | - Mark S. Shiroishi
- Mark and Mary Stevens Neuroimaging and Informatics InstituteKeck School of Medicine, University of Southern CaliforniaLos AngelesCA
- Department of RadiologyKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCA
| | | | - Surjo R. Soekadar
- Clinical Neurotechnology LaboratoryDepartment of Psychiatry and Neurosciences (CCM)Charité ‐ Universitätsmedizin BerlinBerlinGermany
| | | | - Myriam Taga
- NYU Langone Department of NeurologyNew YorkNY
| | - Wai Kwong Tang
- Department of PsychiatryThe Chinese University of Hong KongChina
| | - Gregory T. Thielman
- Department of Physical Therapy and NeuroscienceUniversity of the SciencesPhiladelphiaPA
| | - Daniela Vecchio
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Nick S. Ward
- University College London Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Lars T. Westlye
- Department of PsychologyUniversity of OsloNorway
- Department of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Emilio Werden
- The Florey Institute of Neuroscience and Mental HealthHeidelbergVictoriaAustralia
- Melbourne Dementia Research CenterUniversity of MelbourneVictoriaAustralia
| | - Carolee Winstein
- Biokinesiology and Physical TherapyUniversity of Southern CaliforniaLos AngelesCA
| | - George F. Wittenberg
- Department of NeurologyUniversity of PittsburghPA
- Department of Veterans AffairsGeriatrics Research Educational & Clinical CenterVeterans Affairs Pittsburgh Healthcare System (VAPHS)PittsburghPA
| | - Steven L. Wolf
- Division of Physical TherapyDepartment of Rehabilitation MedicineEmory University School of MedicineAtlantaGA
- Department of MedicineEmory University School of MedicineAtlantaGA
| | - Kristin A. Wong
- Department of Physical Medicine & RehabilitationDell Medical SchoolUniversity of Texas at AustinTX
| | - Chunshui Yu
- Department of RadiologyTianjin Medical University General HospitalTianjinChina
| | - Amy Brodtmann
- The Florey Institute of Neuroscience and Mental HealthHeidelbergVictoriaAustralia
| | - Steven C. Cramer
- Department of NeurologyUniversity of California Los AngelesDavid Geffen School of MedicineLos AngelesCA
- California Rehabilitation HospitalLos AngelesCA
| | - Paul M. Thompson
- Mark and Mary Stevens Neuroimaging and Informatics InstituteKeck School of Medicine, University of Southern CaliforniaLos AngelesCA
| | - Sook‐Lei Liew
- Mark and Mary Stevens Neuroimaging and Informatics InstituteKeck School of Medicine, University of Southern CaliforniaLos AngelesCA
- Chan Division of Occupational Science and Occupational TherapyUniversity of Southern CaliforniaLos AngelesCA
- Biokinesiology and Physical TherapyUniversity of Southern CaliforniaLos AngelesCA
| |
Collapse
|
22
|
Johnson AC, Tremble SM, Cipolla MJ. Experimental Preeclampsia Causes Long-Lasting Hippocampal Vascular Dysfunction and Memory Impairment. Front Physiol 2022; 13:889918. [PMID: 35615682 PMCID: PMC9124928 DOI: 10.3389/fphys.2022.889918] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Preeclampsia (PE) is a hypertensive disorder of pregnancy that is associated with memory impairment, cognitive decline and brain atrophy later in life in women at ages as young as early-to-mid 40 s. PE increases the risk of vascular dementia three-fold, however, long-lasting effects of PE on the vasculature of vulnerable brain regions involved in memory and cognition, such as the hippocampus, remain unknown. Here, we used a rat model of experimental PE (ePE) induced by maintaining rats on a 2% cholesterol diet beginning on day 7 of gestation to investigate hippocampal function later in life. Hippocampal-dependent memory and hippocampal arteriole (HA) function were determined in Sprague Dawley rats 5 months after either a healthy pregnancy or ePE (n = 8/group). Rats that had ePE were hypertensive and had impaired vasoreactivity of HAs to mediators involved in matching neuronal activity with local blood flow (i.e., neurovascular coupling). ePE rats also had impaired long-term memory, but not spatial memory. Thus, this model of ePE mimics some of the long-lasting cardiovascular and cognitive consequences that occur in women who previously had PE. These findings suggest endothelial and vascular smooth muscle dysfunction of HAs were present months after PE that could impair hippocampal neurovascular coupling. This represents a novel vascular mechanism by which PE causes early-onset dementia.
Collapse
Affiliation(s)
- Abbie C. Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, United States,*Correspondence: Abbie C. Johnson,
| | - Sarah M. Tremble
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Marilyn J. Cipolla
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, United States,Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont Larner College of Medicine, Burlington, VT, United States,Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, United States,Department of Electrical and Biomedical Engineering, University of Vermont College of Engineering and Mathematical Sciences, Burlington, VT, United States
| |
Collapse
|
23
|
Unveiling OASIS family as a key player in hypoxia-ischemia cases induced by cocaine using generative adversarial networks. Sci Rep 2022; 12:6734. [PMID: 35469040 PMCID: PMC9038918 DOI: 10.1038/s41598-022-10772-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Repeated cocaine use poses many serious health risks to users. One of the risks is hypoxia and ischemia (HI). To restore the biological system against HI, complex biological mechanisms operate at the gene level. Despite the complexity of biological mechanisms, there are common denominator genes that play pivotal roles in various defense systems. Among these genes, the cAMP response element-binding (Creb) protein contributes not only to various aspects of drug-seeking behavior and drug reward, but also to protective mechanisms. However, it is still unclear which Creb members are key players in the protection of cocaine-induced HI conditions. Herein, using one of the state-of-the-art deep learning methods, the generative adversarial network, we revealed that the OASIS family, one of the Creb family, is a key player in various defense mechanisms such as angiogenesis and unfolded protein response against the HI state by unveiling hidden mRNA expression profiles. Furthermore, we identified mysterious kinases in the OASIS family and are able to explain why the prefrontal cortex and hippocampus are vulnerable to HI at the genetic level.
Collapse
|
24
|
Castro CC, Souza Pagnussat A, Munhoz CD, Netto CA. Coumestrol pre‐treatment improves spatial learning and memory deficits following transient cerebral ischemia recruiting hippocampal
GluR2 AMPA
receptors. Hippocampus 2022; 32:413-418. [DOI: 10.1002/hipo.23418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Cibele Canal Castro
- Department of Biochemistry Institute for Basic Health Sciences, Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Aline Souza Pagnussat
- Department of Physiotherapy Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
| | - Carolina Demarchi Munhoz
- Department of Pharmacology Universidade de São Paulo. Instituto de Ciências Biomedicas São Paulo Brazil
| | - Carlos Alexandre Netto
- Department of Biochemistry Institute for Basic Health Sciences, Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| |
Collapse
|
25
|
Intraarterial anti-leptin therapy via ICA protects ipsilateral CA1 neurons subjected to ischemia and reperfusion. PLoS One 2022; 17:e0261644. [PMID: 35015765 PMCID: PMC8752009 DOI: 10.1371/journal.pone.0261644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/07/2021] [Indexed: 11/19/2022] Open
Abstract
Background Brain reperfusion following an ischemic event is essential for tissue viability, however, it also involves processes that promote neuronal cell death. We have recently shown that local expression of the hormone leptin in cardiovascular organs drives deleterious remodeling. As cerebral ischemia-reperfusion (IR) lesions derive expression of both the leptin hormone and its receptor, we hypothesized that blocking leptin activity in the injured brain area will reduce the deleterious effects of IR injury. Methods C57BL6 male mice underwent bilateral common carotid artery and external carotid artery ligation. The right hemisphere was reperfused after 12 minutes, followed by intraarterial injection of either a low-dose leptin antagonist or saline solution via the ipsilateral ICA. The left common carotid artery remained ligated. Fifteen IR/leptin antagonist-injected and fourteen IR/saline-injected mice completed the experiment. Five days after surgery brains were collected and samples of the hippocampal CA1 region were analyzed for cell viability (H&E) and apoptosis (TUNEL and caspase3), for neuroinflammation (Iba1), and for signaling pathways of pSTAT3 and pSmad2. Results The right hemisphere hippocampal CA1 region subjected to IR and saline injection exhibited increased apoptosis and necrosis of pyramidal cells. Also, increased density of activated microglia/macrophages was evident around the CA1 region. Comparatively, leptin antagonist treatment at reperfusion reduced apoptosis and necrosis of pyramidal cells, as indicated by increased number of viable cells (p < 0.01), and reduced TUNEL (p < 0.001) and caspase3-positive cells (p<0.05). Furthermore, this treatment reduced the density of activated microglia/macrophages (p < 0.001) in the CA1 region. Signaling pathway analysis revealed that while pSTAT3 and pSmad2-positive cells were found surrounding the stratum pyramidal in saline-treated animals, pSTAT3 signal was undetected and pSmad2 was greatly reduced in this territory following leptin antagonist treatment (p < 0.01). Conclusions Inhibition of leptin activity in hemispheric IR injury preserved the viability of ipsilateral hippocampal CA1 neurons, likely by preventing apoptosis and local inflammation. These results indicate that intraarterial anti-leptin therapy may have clinical potential in reducing hemispheric brain IR injury.
Collapse
|
26
|
Wang S, Jiao F, Border JJ, Fang X, Crumpler RF, Liu Y, Zhang H, Jefferson J, Guo Y, Elliott PS, Thomas KN, Strong LB, Urvina AH, Zheng B, Rijal A, Smith SV, Yu H, Roman RJ, Fan F. Luseogliflozin, a sodium-glucose cotransporter-2 inhibitor, reverses cerebrovascular dysfunction and cognitive impairments in 18-mo-old diabetic animals. Am J Physiol Heart Circ Physiol 2022; 322:H246-H259. [PMID: 34951541 PMCID: PMC8759958 DOI: 10.1152/ajpheart.00438.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/29/2021] [Accepted: 12/15/2021] [Indexed: 02/03/2023]
Abstract
Diabetes mellitus (DM) is a leading risk factor for age-related dementia, but the mechanisms involved are not well understood. We previously discovered that hyperglycemia induced impaired myogenic response (MR) and cerebral blood flow (CBF) autoregulation in 18-mo-old DM rats associated with blood-brain barrier (BBB) leakage, impaired neurovascular coupling, and cognitive impairment. In the present study, we examined whether reducing plasma glucose with a sodium-glucose cotransporter-2 inhibitor (SGLT2i) luseogliflozin can ameliorate cerebral vascular and cognitive function in diabetic rats. Plasma glucose and HbA1c levels of 18-mo-old DM rats were reduced, and blood pressure was not altered after treatment with luseogliflozin. SGLT2i treatment restored the impaired MR of middle cerebral arteries (MCAs) and parenchymal arterioles and surface and deep cortical CBF autoregulation in DM rats. Luseogliflozin treatment also rescued neurovascular uncoupling, reduced BBB leakage and cognitive deficits in DM rats. However, SGLT2i did not have direct constrictive effects on vascular smooth muscle cells and MCAs isolated from normal rats, although it decreased reactive oxygen species production in cerebral vessels of DM rats. These results provide evidence that normalization of hyperglycemia with an SGLT2i can reverse cerebrovascular dysfunction and cognitive impairments in rats with long-standing hyperglycemia, possibly by ameliorating oxidative stress-caused vascular damage.NEW & NOTEWORTHY This study demonstrates that luseogliflozin, a sodium-glucose cotransporter-2 inhibitor, improved CBF autoregulation in association with reduced vascular oxidative stress and AGEs production in the cerebrovasculature of 18-mo-old DM rats. SGLT2i also prevented BBB leakage, impaired functional hyperemia, neurodegeneration, and cognitive impairment seen in DM rats. Luseogliflozin did not have direct constrictive effects on VSMCs and MCAs isolated from normal rats. These results provide evidence that normalization of hyperglycemia with an SGLT2i can reverse cerebrovascular dysfunction and cognitive impairments in rats with long-standing hyperglycemia, possibly by ameliorating oxidative stress-caused vascular damage.
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Feng Jiao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jane J Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Reece F Crumpler
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joshua Jefferson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ya Guo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Parker S Elliott
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kirby N Thomas
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Luke B Strong
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Austin H Urvina
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Baoying Zheng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Arjun Rijal
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Stanley V Smith
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
27
|
Khlif MS, Werden E, Bird LJ, Egorova-Brumley N, Brodtmann A. Atrophy of Ipsilesional Hippocampal Subfields Vary Over First Year After Ischemic Stroke. J Magn Reson Imaging 2021; 56:273-281. [PMID: 34837426 DOI: 10.1002/jmri.28009] [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: 10/05/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND The structural integrity of hippocampal subfields has been investigated in many neurological disorders and was shown to be better associated with cognitive performance than whole hippocampus. In stroke, hippocampal atrophy is linked to cognitive impairment, but it is unknown whether the hippocampal subfields atrophy differently. PURPOSE To evaluate longitudinal hippocampal subfield atrophy in first year poststroke, in comparison with atrophy in healthy individuals. STUDY TYPE Cohort. SUBJECTS A total of 92 ischemic stroke (age: 67 ± 12 years, 63 men) and 39 healthy participants (age: 69 ± 7 years, 24 men). FIELD STRENGTH/SEQUENCE A3 T/T1-MPRAGE, T2-SPACE, and T2-FLAIR. ASSESSMENT FreeSurfer (6.0) was used to delineate 12 hippocampal subfields. Whole hippocampal volume was computed as sum of subfield volumes excluding hippocampal fissure volume. Separate assessments were completed for contralesional and ipsilesional hippocampi. STATISTICAL TESTS A mixed-effect regression model was used to compare subfield volumes cross-sectionally between healthy and stroke groups and longitudinally between 3-month and 12-month timepoints. False discovery rate at 0.05 significance level was used to correct for multiple comparisons. Also, a receiver operating characteristic (ROC) curve analysis was performed to assess differentiation between healthy and stroke participants based on subfield volumes. RESULTS There were no volume differences between groups at 3 months, but there was a significant difference (P = 0.027) in whole hippocampal volume reduction over time between control and stroke ipsilesionally. Thus, the ipsilesional whole hippocampal volume in stroke became significantly smaller (P = 0.035) at 12 months. The hippocampal tail was the highest single-region contributor (22.7%) to ipsilesional hippocampal atrophy (1.19%) over 9 months. The cornu ammonis areas (CA1) subfield volume reduction was minimal in controls and stroke contralesionally but significant ipsilesionally (P = 0.007). CA1 volume significantly outperformed whole hippocampal volume (P < 0.01) in discriminating between stroke participants and healthy controls in ROC curve analysis. DATA CONCLUSION Greater stroke-induced effects were observed in the ipsilesional hippocampus anteriorly in CA1 and posteriorly in the hippocampal tail. Atrophy of CA1 and hippocampal tail may provide a better link to cognitive impairment than whole hippocampal atrophy. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY STAGE: 3.
Collapse
Affiliation(s)
- Mohamed Salah Khlif
- The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Emilio Werden
- The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Laura J Bird
- The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Natalia Egorova-Brumley
- The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia.,Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Australia
| | - Amy Brodtmann
- The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia.,Department of Neurology, Austin Health, Heidelberg, Victoria, Australia.,Department of Neurology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Eastern Cognitive Disorders Clinic, Box Hill Hospital, Monash University, Box Hill, Victoria, Australia
| |
Collapse
|
28
|
Li Z, Xu H, Xu Y, Lu G, Peng Q, Chen J, Bi R, Li J, Chen S, Li H, Jin H, Hu B. Morinda officinalis oligosaccharides alleviate depressive-like behaviors in post-stroke rats via suppressing NLRP3 inflammasome to inhibit hippocampal inflammation. CNS Neurosci Ther 2021; 27:1570-1586. [PMID: 34559953 PMCID: PMC8611777 DOI: 10.1111/cns.13732] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/11/2021] [Accepted: 09/11/2021] [Indexed: 01/14/2023] Open
Abstract
Aims Morinda officinalis oligosaccharides (MOOs), a traditional Chinese medicine, have been used to treat mild and moderate depressive episodes. In this study, we investigated whether MOOs can ameliorate depressive‐like behaviors in post‐stroke depression (PSD) rats and further explored its mechanism by suppressing microglial NLRP3 inflammasome activation to inhibit hippocampal inflammation. Methods Behavioral tests were performed to evaluate the effect of MOOs on depressive‐like behaviors in PSD rats. The effects of MOOs on the expression of IL‐18, IL‐1β, and nucleotide‐binding domain leucine‐rich repeat (NLR) family pyrin domain containing 3 (NLRP3) inflammasome were measured in both PSD rats and lipopolysaccharide (LPS) and adenosine triphosphate (ATP) stimulated primary rat microglia by reverse transcription polymerase chain reaction (RT‐PCR), immunofluorescence and Western blot analysis. Adeno‐associated virus (AAV) was injected into the hippocampus to regulate NLRP3 inflammasome expression. The detailed molecular mechanism underlying the effects of MOOs was analyzed by Western blot and immunofluorescence. Results MOOs can alleviate depressive‐like behaviors in PSD rats. PSD rats showed increased expression of IL‐18, IL‐1β, and NLRP3 inflammasome in the ischemic hippocampus, while MOOs reversed the elevation. NLRP3 downregulation ameliorated depressive‐like behaviors and hippocampal inflammation response in PSD rats, while NLRP3 upregulation inhibited the effect of MOOs on depressive‐like behaviors and hippocampal inflammation response in PSD rats. Moreover, we found that NLRP3 was mainly expressed on microglia. In vitro, MOOs effectively inhibited the expression of IL‐18, IL‐1β, and NLRP3 inflammasome in LPS + ATP treated primary rat microglia. We also showed that modulation of NLRP3 inflammasome by MOOs was associated with the IκB/NF‐κB p65 signaling pathway. Conclusion Overall, our study reveals the antidepressive effect of MOOs on PSD rats through modulation of microglial NLRP3 inflammasome. We also provide a novel insight into hippocampal inflammation response in PSD pathology and put forward NLRP3 inflammasome as a potential therapeutic target for PSD.
Collapse
Affiliation(s)
- Zhifang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hexiang Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Xu
- Institute of Science, Beijing Tongrentang Co., Ltd., Beijing, China
| | - Guanfeng Lu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiwei Peng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiefang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianzhuang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengcai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongkai Li
- Institute of Science, Beijing Tongrentang Co., Ltd., Beijing, China
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Yang YS, Choi JH, Rah JC. Hypoxia with inflammation and reperfusion alters membrane resistance by dynamically regulating voltage-gated potassium channels in hippocampal CA1 neurons. Mol Brain 2021; 14:147. [PMID: 34556177 PMCID: PMC8461870 DOI: 10.1186/s13041-021-00857-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/07/2021] [Indexed: 02/01/2023] Open
Abstract
Hypoxia typically accompanies acute inflammatory responses in patients and animal models. However, a limited number of studies have examined the effect of hypoxia in combination with inflammation (Hypo-Inf) on neural function. We previously reported that neuronal excitability in hippocampal CA1 neurons decreased during hypoxia and greatly rebounded upon reoxygenation. We attributed this altered excitability mainly to the dynamic regulation of hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels and input resistance. However, the molecular mechanisms underlying input resistance changes by Hypo-Inf and reperfusion remained unclear. In the present study, we found that a change in the density of the delayed rectifier potassium current (IDR) can explain the input resistance variability. Furthermore, voltage-dependent inactivation of A-type potassium (IA) channels shifted in the depolarizing direction during Hypo-Inf and reverted to normal upon reperfusion without a significant alteration in the maximum current density. Our results indicate that changes in the input resistance, and consequently excitability, caused by Hypo-Inf and reperfusion are at least partially regulated by the availability and voltage dependence of KV channels. Moreover, these results suggest that selective KV channel modulators can be used as potential neuroprotective drugs to minimize hypoxia- and reperfusion-induced neuronal damage.
Collapse
Affiliation(s)
- Yoon-Sil Yang
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
| | - Joon Ho Choi
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
| | - Jong-Cheol Rah
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333 Techno Jungang-daero, Dalseong-gun, Daegu, 42988 South Korea
| |
Collapse
|
30
|
Zhang G, Lai Z, Gu L, Xu K, Wang Z, Duan Y, Chen H, Zhang M, Zhang J, Zhao Z, Wang S. Delta Opioid Receptor Activation with Delta Opioid Peptide [d-Ala2, d-Leu5] Enkephalin Contributes to Synaptic Improvement in Rat Hippocampus against Global Ischemia. Cell Transplant 2021; 30:9636897211041585. [PMID: 34470528 PMCID: PMC8419564 DOI: 10.1177/09636897211041585] [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] [Indexed: 11/28/2022] Open
Abstract
Global cerebral ischemia induced by cardiac arrest usually leads to poor neurological outcomes. Numerous studies have focused on ways to prevent ischemic damage in the brain, however clinical therapies are still limited. Our previous studies revealed that delta opioid receptor (DOR) activation with [d-Ala2, d-Leu5] enkephalin (DADLE), a DOR agonist, not only significantly promotes neuronal survival on day 3, but also improves spatial memory deficits on days 5-9 after ischemia. However, the neurological mechanism underlying DADLE-induced cognitive recovery remains unclear. This study first examined the changes in neuronal survival in the CA1 region at the advanced time point (day 7) after ischemia/reperfusion (I/R) injury and found a significant amelioration of damaged CA1 neurons in the rats treated with DADLE (2.5 nmol) when administered at the onset of reperfusion. The structure and function of CA1 neurons on days 3 and 7 post-ischemia showed significant improvements in both the density of the injured dendritic spines and the basic transmission of the impaired CA3-CA1 synapses following DADLE treatment. The molecular changes involved in DADLE-mediated synaptic modulation on days 3 and 7 post-ischemia implied the time-related differential regulation of PKCα-MARCKS on the dendritic spine structure and of BDNF- ERK1/2-synapsin I on synaptic function, in response to ischemic/reperfusion injury as well as to DADLE treatment. Importantly, all the beneficial effects of DADLE on ischemia-induced cellular, synaptic, and molecular deficits were eliminated by the DOR inhibitor naltrindole (2.5 nmol). Taken together, this study suggested that DOR activation-induced protective signaling pathways of PKCα-MARCKS involved in the synaptic morphology and BDNF-ERK-synapsin I in synaptic transmission may be engaged in the cognitive recovery in rats suffering from advanced cerebral ischemia.
Collapse
Affiliation(s)
- Guangming Zhang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Zelin Lai
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Lingling Gu
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Kejia Xu
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Zhenlu Wang
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Yale Duan
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Huifen Chen
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital
| | - Min Zhang
- Tongji University School of Medicine, Shanghai 201204, China
| | - Jun Zhang
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital.,Tongji University School of Medicine, Shanghai 201204, China
| | - Zheng Zhao
- Shanghai Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Shuyan Wang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
31
|
Faillot M, Chaillet A, Palfi S, Senova S. Rodent models used in preclinical studies of deep brain stimulation to rescue memory deficits. Neurosci Biobehav Rev 2021; 130:410-432. [PMID: 34437937 DOI: 10.1016/j.neubiorev.2021.08.012] [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: 02/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Deep brain stimulation paradigms might be used to treat memory disorders in patients with stroke or traumatic brain injury. However, proof of concept studies in animal models are needed before clinical translation. We propose here a comprehensive review of rodent models for Traumatic Brain Injury and Stroke. We systematically review the histological, behavioral and electrophysiological features of each model and identify those that are the most relevant for translational research.
Collapse
Affiliation(s)
- Matthieu Faillot
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Antoine Chaillet
- Laboratoire des Signaux et Systèmes (L2S-UMR8506) - CentraleSupélec, Université Paris Saclay, Institut Universitaire de France, France
| | - Stéphane Palfi
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Suhan Senova
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France.
| |
Collapse
|
32
|
Protective Role of Glutathione in the Hippocampus after Brain Ischemia. Int J Mol Sci 2021; 22:ijms22157765. [PMID: 34360532 PMCID: PMC8345998 DOI: 10.3390/ijms22157765] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
Stroke is a major cause of death worldwide, leading to serious disability. Post-ischemic injury, especially in the cerebral ischemia-prone hippocampus, is a serious problem, as it contributes to vascular dementia. Many studies have shown that in the hippocampus, ischemia/reperfusion induces neuronal death through oxidative stress and neuronal zinc (Zn2+) dyshomeostasis. Glutathione (GSH) plays an important role in protecting neurons against oxidative stress as a major intracellular antioxidant. In addition, the thiol group of GSH can function as a principal Zn2+ chelator for the maintenance of Zn2+ homeostasis in neurons. These lines of evidence suggest that neuronal GSH levels could be a key factor in post-stroke neuronal survival. In neurons, excitatory amino acid carrier 1 (EAAC1) is involved in the influx of cysteine, and intracellular cysteine is the rate-limiting substrate for the synthesis of GSH. Recently, several studies have indicated that cysteine uptake through EAAC1 suppresses ischemia-induced neuronal death via the promotion of hippocampal GSH synthesis in ischemic animal models. In this article, we aimed to review and describe the role of GSH in hippocampal neuroprotection after ischemia/reperfusion, focusing on EAAC1.
Collapse
|
33
|
Yan N, Xu Z, Qu C, Zhang J. Dimethyl fumarate improves cognitive deficits in chronic cerebral hypoperfusion rats by alleviating inflammation, oxidative stress, and ferroptosis via NRF2/ARE/NF-κB signal pathway. Int Immunopharmacol 2021; 98:107844. [PMID: 34153667 DOI: 10.1016/j.intimp.2021.107844] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
Cerebrovascular disease and its risk factors cause persistent decrease of cerebral blood flow, chronic cerebral hypoperfusion (CCH) is the major foundation of vascular cognitive impairment (VCI). The hippocampus is extremely vulnerable to cerebral ischemia and hypoxia. Oxidative stress and neuroinflammation injury are important pathophysiological mechanisms of this process, which is closely related to hippocampal neurons damage and loss. Dimethyl fumarate (DMF), an FDA-approved therapeutic for multiple sclerosis (MS), plays a protective role in multiple neurological disorders. Studies have shown that DMF exerts anti-inflammatory and antioxidant effects via the NRF2/ARE/NF-κB signaling pathway. Thus, this study aimed to evaluate the neuroprotective effect of DMF in the CCH rat model. Ferroptosis, a novel defined iron-dependent cell death form, were found to be strongly associated with the pathophysiology of CCH. Emerging evidences have shown that inhibition of ferroptosis by targeting NRF2 exerted neuroprotective effect in neurodegeneration diseases. We also investigated whether DMF can alleviate cognitive deficits through inhibition of ferroptosis by the NRF2 signaling pathway in this study. DMF was intragastric for consecutive five weeks (100 mg/kg/day). Then behavior test and histological, molecular, and biochemical analysis were performed. We found that DMF treatment significantly improved cognitive deficits and partially reversed hippocampus neuronal damage and loss caused by CCH. And DMF treatment decreased hippocampus IL-1β, TNF-α, and IL-6 pro-inflammatory cytokines concentration, and mediated the NF-κB signaling pathway. And DMF also alleviated hippocampus oxidative stress through reducing MDA, and increasing GSH and SOD levels, which are also closely associated with ferroptosis. Besides, DMF treatment reduced the expression of PTGS2, and increased the expression of FTH1 and xCT, and the iron content is also reduced, which were the important features related to ferroptosis. Furthermore, DMF activated the NRF2/ARE signaling pathway and upregulated the expression of HO-1, NQO1 and GPX4. These outcomes indicated that DMF can improve cognitive impairment in rats with CCH, possibly through alleviating neuroinflammation, oxidative stress damage and inhibiting ferroptosis of hippocampal neurons. Overall, our results provide new evidence for the neuroprotective role of DMF.
Collapse
Affiliation(s)
- Nao Yan
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - Zhipeng Xu
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - Changhua Qu
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - JunJian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China.
| |
Collapse
|
34
|
Khlif MS, Bird LJ, Restrepo C, Khan W, Werden E, Egorova‐Brumley N, Brodtmann A. Hippocampal subfield volumes are associated with verbal memory after first-ever ischemic stroke. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12195. [PMID: 34136634 PMCID: PMC8197170 DOI: 10.1002/dad2.12195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Hippocampal subfield volumes are more closely associated with cognitive impairment than whole hippocampal volume in many diseases. Both memory and whole hippocampal volume decline after stroke. Understanding the subfields' temporal evolution could reveal valuable information about post-stroke memory. METHODS We sampled 120 participants (38 control, 82 stroke), with cognitive testing and 3T-MRI available at 3 months and 3 years, from the Cognition and Neocortical Volume after Stroke (CANVAS) study. Verbal memory was assessed using the Hopkins Verbal Learning Test-Revised. Subfields were delineated using FreeSurfer. We used partial Pearson's correlation to assess the associations between subfield volumes and verbal memory scores, adjusting for years of education, sex, and stroke side. RESULTS The left cornu ammonis areas 2/3 and hippocampal tail volumes were significantly associated with verbal memory 3-month post-stroke. At 3 years, the associations became stronger and involved more subfields. DISCUSSION Hippocampal subfield volumes may be a useful biomarker for post-stroke cognitive impairment.
Collapse
Affiliation(s)
- Mohamed Salah Khlif
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Laura J. Bird
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Carolina Restrepo
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Wasim Khan
- Department of NeuroscienceCentral Clinical SchoolMonash UniversityClaytonVictoriaAustralia
- Department of Neuroimaging Institute of PsychiatryPsychology, and Neuroscience (IoPPN), King's College LondonLondonUK
| | - Emilio Werden
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Natalia Egorova‐Brumley
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Melbourne School of Psychological SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Amy Brodtmann
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAustin HealthHeidelbergVictoriaAustralia
- Eastern Cognitive Disorders ClinicBox Hill HospitalMonash UniversityBox HillVictoriaAustralia
| |
Collapse
|
35
|
González Fuentes J, Insausti Serrano R, Cebada Sánchez S, Lagartos Donate MJ, Rivas Infante E, Arroyo Jiménez MDM, Marcos Rabal MDP. Neuropeptides in the developing human hippocampus under hypoxic-ischemic conditions. J Anat 2021; 239:856-868. [PMID: 34028021 PMCID: PMC8450465 DOI: 10.1111/joa.13458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 11/26/2022] Open
Abstract
The perinatal period, sensitive for newborn survival, is also one of the most critical moments in human brain development. Perinatal hypoxia due to reduced blood supply to the brain (ischemia) is one of the main causes of neonatal mortality. Brain damage caused by perinatal hypoxia–ischemia (HI) can lead to neuro‐ and psychological disorders. However, its impact seems to be region‐dependent, with the hippocampus being one of the most affected areas. Among the neuronal populations of the hippocampus, some interneuron groups – such as somatostatin‐ or neuropeptide Y‐expressing neurons – seem to be particularly vulnerable. The limited information available about the effects of HI in the hippocampus comes mainly from animal models and adult human studies. This article presents an immunohistochemical analysis of somatostatin (SOM) and neuropeptide Y (NPY) expression in the developing human hippocampus after perinatal HI. Two rostrocaudal sections of the body of the hippocampus were analysed, and the number of immunostained cells in the polymorphic layer of the dentate gyrus (DG) and the pyramidal cell layer and stratumoriens of the CA3, CA2 and CA1 fields of the hippocampus proper were quantified. The results showed a lower density of both neuropeptides in hypoxic compared to control cases. In the HI group, the number of SOM‐immunoreactive cell bodies was statistically significantly lower in the pyramidal cell layer and stratumoriens of CA1, while the number of NPY‐expressing neurons was statistically lower in the pyramidal cell layer of CA2. Besides, the number of SOM‐expressing neurons was significantly higher in the stratumoriens of CA1 compared to that in CA2. In sum, we observed a different vulnerability of SOM‐ and NPY‐containing neurons in the developing human hippocampus following perinatal HI damage. Our results could contribute to a better understanding of the behaviour of these neuronal populations under stressful conditions during the perinatal period.
Collapse
Affiliation(s)
- Joaquín González Fuentes
- Cellular Neuroanatomy and Molecular Chemistry of Central Nervous System, School of Pharmacy and School of Medicine, University of Castilla-La Mancha (UCLM), Centro Regional de Investigaciones Biomédicas, Albacete, Spain
| | | | | | | | | | - María Del Mar Arroyo Jiménez
- Cellular Neuroanatomy and Molecular Chemistry of Central Nervous System, School of Pharmacy and School of Medicine, University of Castilla-La Mancha (UCLM), Centro Regional de Investigaciones Biomédicas, Albacete, Spain
| | - María Del Pilar Marcos Rabal
- Cellular Neuroanatomy and Molecular Chemistry of Central Nervous System, School of Pharmacy and School of Medicine, University of Castilla-La Mancha (UCLM), Centro Regional de Investigaciones Biomédicas, Albacete, Spain
| |
Collapse
|
36
|
Environmental Enrichment Enhances Ca v 2.1 Channel-Mediated Presynaptic Plasticity in Hypoxic-Ischemic Encephalopathy. Int J Mol Sci 2021; 22:ijms22073414. [PMID: 33810296 PMCID: PMC8037860 DOI: 10.3390/ijms22073414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Hypoxic–ischemic encephalopathy (HIE) is a devastating neonatal brain condition caused by lack of oxygen and limited blood flow. Environmental enrichment (EE) is a classic paradigm with a complex stimulation of physical, cognitive, and social components. EE can exert neuroplasticity and neuroprotective effects in immature brains. However, the exact mechanism of EE on the chronic condition of HIE remains unclear. HIE was induced by a permanent ligation of the right carotid artery, followed by an 8% O2 hypoxic condition for 1 h. At 6 weeks of age, HIE mice were randomly assigned to either standard cages or EE cages. In the behavioral assessments, EE mice showed significantly improved motor performances in rotarod tests, ladder walking tests, and hanging wire tests, compared with HIE control mice. EE mice also significantly enhanced cognitive performances in Y-maze tests. Particularly, EE mice showed a significant increase in Cav 2.1 (P/Q type) and presynaptic proteins by molecular assessments, and a significant increase of Cav 2.1 in histological assessments of the cerebral cortex and hippocampus. These results indicate that EE can upregulate the expression of the Cav 2.1 channel and presynaptic proteins related to the synaptic vesicle cycle and neurotransmitter release, which may be responsible for motor and cognitive improvements in HIE.
Collapse
|
37
|
Pires G, Leitner D, Drummond E, Kanshin E, Nayak S, Askenazi M, Faustin A, Friedman D, Debure L, Ueberheide B, Wisniewski T, Devinsky O. Proteomic differences in the hippocampus and cortex of epilepsy brain tissue. Brain Commun 2021; 3:fcab021. [PMID: 34159317 DOI: 10.1093/braincomms/fcab021] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/22/2022] Open
Abstract
Epilepsy is a common neurological disorder affecting over 70 million people worldwide, with a high rate of pharmaco-resistance, diverse comorbidities including progressive cognitive and behavioural disorders, and increased mortality from direct (e.g. sudden unexpected death in epilepsy, accidents, drowning) or indirect effects of seizures and therapies. Extensive research with animal models and human studies provides limited insights into the mechanisms underlying seizures and epileptogenesis, and these have not translated into significant reductions in pharmaco-resistance, morbidities or mortality. To help define changes in molecular signalling networks associated with seizures in epilepsy with a broad range of aetiologies, we examined the proteome of brain samples from epilepsy and control cases. Label-free quantitative mass spectrometry was performed on the hippocampal cornu ammonis 1-3 region (CA1-3), frontal cortex and dentate gyrus microdissected from epilepsy and control cases (n = 14/group). Epilepsy cases had significant differences in the expression of 777 proteins in the hippocampal CA1 - 3 region, 296 proteins in the frontal cortex and 49 proteins in the dentate gyrus in comparison to control cases. Network analysis showed that proteins involved in protein synthesis, mitochondrial function, G-protein signalling and synaptic plasticity were particularly altered in epilepsy. While protein differences were most pronounced in the hippocampus, similar changes were observed in other brain regions indicating broad proteomic abnormalities in epilepsy. Among the most significantly altered proteins, G-protein subunit beta 1 (GNB1) was one of the most significantly decreased proteins in epilepsy in all regions studied, highlighting the importance of G-protein subunit signalling and G-protein-coupled receptors in epilepsy. Our results provide insights into common molecular mechanisms underlying epilepsy across various aetiologies, which may allow for novel targeted therapeutic strategies.
Collapse
Affiliation(s)
- Geoffrey Pires
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA.,Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA.,Alzheimer's and Prion Diseases Team, Paris Brain Institute, CNRS, UMR 7225, INSERM 1127, Sorbonne University UM75, Paris, France
| | - Dominique Leitner
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Eleanor Drummond
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA.,Faculty of Medicine and Health, Brain and Mind Centre and School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Shruti Nayak
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Manor Askenazi
- Biomedical Hosting LLC, USA.,Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Arline Faustin
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Daniel Friedman
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ludovic Debure
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Beatrix Ueberheide
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA.,Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA.,Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, NY, USA.,Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.,Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
38
|
Stevenson W, Hase Y, Wilson E, Hollins A, Hase M, Ennaceur A, Craggs L, Ihara M, Horsburgh K, Kalaria RN. Long-term effects of experimental carotid stenosis on hippocampal infarct pathology, neurons and glia and amelioration by environmental enrichment. Brain Res Bull 2020; 163:72-83. [PMID: 32707262 DOI: 10.1016/j.brainresbull.2020.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/03/2020] [Accepted: 07/15/2020] [Indexed: 01/17/2023]
Abstract
Hippocampal atrophy and pathology are common in ageing-related disorders and associated with cognitive impairment and dementia. We explored whether environmental enrichment (EE) ameliorated the pathological sequelae in the hippocampus subsequent to chronic cerebral hypoperfusion induced by bilateral common carotid artery stenosis (BCAS). Seventy-four male C57BL/6 J mice underwent BCAS or sham surgery. One-week after surgery, mice were exposed to three different degrees of EE; either standard housing conditions (std), limited 3 -h exposure to EE per day (3 h) or full-time exposure to EE (full) for 3 months. Four months after surgery, the hippocampus was examined for the extent of vascular brain injury and neuronal and glial changes. Results showed that long-term BCAS induced strokes, most often in CA1 subfield, reduced 40-50 % CA1 neurons (P < 0.01) and increased microglia/macrophage in CA1-CA3 subfields (P < 0.02). Remarkably, both 3 h and full-time EE regimes attenuated hippocampal neuronal death and repressed recurrent strokes with complete prevention of larger infarcts in mice on full-time EE (P < 0.01). Full-time EE also reduced astrocytic clasmatodendrosis and microglial/macrophage activation in all CA subfields. Our results suggest that exposure to EE differentially reduces long-term hypoperfusive hippocampal damage. The implementation of even limited EE may be beneficial for patients diagnosed with vascular cognitive impairment.
Collapse
Affiliation(s)
- William Stevenson
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Yoshiki Hase
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Elle Wilson
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Annabel Hollins
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mai Hase
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Abdel Ennaceur
- Department of Pharmacy, Sunderland Pharmacy School, The University of Sunderland, Sunderland, UK
| | - Lucy Craggs
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Centre, Osaka, Japan
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Raj N Kalaria
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
39
|
Investigating the Structure of Neurotoxic Protein Aggregates Inside Cells. Trends Cell Biol 2020; 30:951-966. [PMID: 32981805 DOI: 10.1016/j.tcb.2020.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022]
Abstract
Neurodegenerative diseases affect the lives of millions of people across the world, being particularly prevalent in the aging population. Despite huge research efforts, conclusive insights into the disease mechanisms are still lacking. Therefore, therapeutic strategies are limited to symptomatic treatments. A common histopathological hallmark of many neurodegenerative diseases is the presence of large pathognomonic protein aggregates, but their role in the disease pathology is unclear and subject to controversy. Here, we discuss imaging methods allowing investigation of these structures within their cellular environment: conventional electron microscopy (EM), super-resolution light microscopy (SR-LM), and cryo-electron tomography (cryo-ET). Multidisciplinary approaches are key for understanding neurodegenerative diseases and may contribute to the development of effective treatments. For simplicity, we focus on huntingtin aggregates, characteristic of Huntington's disease.
Collapse
|
40
|
León-Moreno LC, Castañeda-Arellano R, Aguilar-García IG, Desentis-Desentis MF, Torres-Anguiano E, Gutiérrez-Almeida CE, Najar-Acosta LJ, Mendizabal-Ruiz G, Ascencio-Piña CR, Dueñas-Jiménez JM, Rivas-Carrillo JD, Dueñas-Jiménez SH. Kinematic Changes in a Mouse Model of Penetrating Hippocampal Injury and Their Recovery After Intranasal Administration of Endometrial Mesenchymal Stem Cell-Derived Extracellular Vesicles. Front Cell Neurosci 2020; 14:579162. [PMID: 33192324 PMCID: PMC7533596 DOI: 10.3389/fncel.2020.579162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022] Open
Abstract
Locomotion speed changes appear following hippocampal injury. We used a hippocampal penetrating brain injury mouse model to analyze other kinematic changes. We found a significant decrease in locomotion speed in both open-field and tunnel walk tests. We described a new quantitative method that allows us to analyze and compare the displacement curves between mice steps. In the tunnel walk, we marked mice with indelible ink on the knee, ankle, and metatarsus of the left and right hindlimbs to evaluate both in every step. Animals with hippocampal damage exhibit slower locomotion speed in both hindlimbs. In contrast, in the cortical injured group, we observed significant speed decrease only in the right hindlimb. We found changes in the displacement patterns after hippocampal injury. Mesenchymal stem cell-derived extracellular vesicles had been used for the treatment of several diseases in animal models. Here, we evaluated the effects of intranasal administration of endometrial mesenchymal stem cell-derived extracellular vesicles on the outcome after the hippocampal injury. We report the presence of vascular endothelial growth factor, granulocyte–macrophage colony-stimulating factor, and interleukin 6 in these vesicles. We observed locomotion speed and displacement pattern preservation in mice after vesicle treatment. These mice had lower pyknotic cells percentage and a smaller damaged area in comparison with the nontreated group, probably due to angiogenesis, wound repair, and inflammation decrease. Our results build up on the evidence of the hippocampal role in walk control and suggest that the extracellular vesicles could confer neuroprotection to the damaged hippocampus.
Collapse
Affiliation(s)
- Lilia Carolina León-Moreno
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico.,Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Rolando Castañeda-Arellano
- Laboratory of Tissue Engineering and Transplant, Department of Physiology, cGMP Cell Processing Facility, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Irene Guadalupe Aguilar-García
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | | | - Elizabeth Torres-Anguiano
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Coral Estefanía Gutiérrez-Almeida
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Luis Jesús Najar-Acosta
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Gerardo Mendizabal-Ruiz
- Department of Computer Sciences, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara, Mexico
| | - César Rodolfo Ascencio-Piña
- Department of Computer Sciences, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara, Mexico
| | - Judith Marcela Dueñas-Jiménez
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Jorge David Rivas-Carrillo
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Sergio Horacio Dueñas-Jiménez
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
41
|
Li Z, Meng X, Ren M, Shao M. Combination of Scalp Acupuncture with Exercise Therapy Effectively Counteracts Ischemic Brain Injury in Rats. J Stroke Cerebrovasc Dis 2020; 29:105286. [PMID: 33066914 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/14/2020] [Accepted: 08/23/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Stroke is one of the leading causes of death and disability worldwide. Scalp acupuncture and exercise therapy have been proven as two effective methods for the treatment of stroke. However, their combined action and mechanisms have not been fully elucidated. The present study aimed to investigate the protective effect of scalp acupuncture combined with exercise therapy on neurons in rats with ischemic brain injury. METHODS 100 rats were randomly divided into 5 groups including sham group, model group, acupuncture group, rehabilitation group, and experimental group (scalp acupuncture combined with exercise therapy). Middle cerebral artery occlusion (MCAO) model in rats was established according to Longa modified suture method to mimic ischemic stroke. The modified Bedexer's neurological function score was used to evaluate the neurological deficits of rats and the brain infarct volume was measured using 2, 3, 5-triphenyl tetrazolium chloride monohydrate (TTC) staining. Moreover, the apoptosis in the hippocampus was detected by western blotting and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The pro-inflammatory cytokines such as interleukin-1 beta (IL-1β) and tumor necrosis factor-alpha (TNF-α), reactive oxygen species (ROS) and superoxide dismutase (SOD) were determined by corresponding kits. Immunohistochemistry or immunofluorescence was performed to detect the expression of brain-derived neurotrophic factor (BDNF), S100β and glial fibrillary acidic protein (GFAP) in the hippocampi of rats. RESULTS The neurological deficit score, the expression levels of apoptotic factors such as cleaved caspase-3 and Bax, and the TUNEL-positive cell rate of the experimental group were significantly lower than those of the acupuncture group and the rehabilitation group. However, apoptosis inhibitor Bcl-2 showed downregulated expression in the MCAO model rats but this trend was reverted by single and combinatorial treatments. In addition, the contents of TNF-α, IL-1β and ROS in the acupuncture group and the rehabilitation group were significantly lower than those in the model group, but higher than the experimental group. While the opposite results were obtained in SOD activity. Furthermore, compared with the model group, the ratios of BDNF, S100β, and GFAP-positive cells in the acupuncture, rehabilitation and experimental groups were significantly increased, and the highest ratios were recorded in the experimental group. CONCLUSIONS This study demonstrated that scalp acupuncture combined with exercise therapy effectively counteracts ischemic brain injury via the downregulation of pro-inflammatory mediators and ROS, the increased production of the antioxidant enzyme SOD, neurotrophic factor BDNF and astrocyte activities.
Collapse
Affiliation(s)
- Zhenjing Li
- Department of Rehabilitation, Pudong New Area People's Hospital affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201299, P. R. China
| | - Xianzhong Meng
- Department of Rehabilitation, Pudong New Area People's Hospital affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201299, P. R. China.
| | - Min Ren
- Department of Rehabilitation, Pudong New Area People's Hospital affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201299, P. R. China
| | - Minglu Shao
- Department of Rehabilitation, Pudong New Area People's Hospital affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201299, P. R. China
| |
Collapse
|
42
|
Experimental Pretreatment with Chlorogenic Acid Prevents Transient Ischemia-Induced Cognitive Decline and Neuronal Damage in the Hippocampus through Anti-Oxidative and Anti-Inflammatory Effects. Molecules 2020; 25:molecules25163578. [PMID: 32781658 PMCID: PMC7463954 DOI: 10.3390/molecules25163578] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022] Open
Abstract
Chlorogenic acid (CGA), an ester of caffeic acid and quinic acid, is among the phenolic acid compounds which can be naturally found in green coffee extract and tea. CGA has been studied since it displays significant pharmacological properties. The aim of this study was to investigate the effects of CGA on cognitive function and neuroprotection including its mechanisms in the hippocampus following transient forebrain ischemia in gerbils. Memory and learning following the ischemia was investigated by eight-arm radial maze and passive avoidance tests. Neuroprotection was examined by immunohistochemistry for neuronal nuclei-specific protein and Fluoro-Jade B histofluorescence staining. For mechanisms of the neuroprotection, alterations in copper, zinc-superoxide dismutase (SOD1), SOD2 as antioxidant enzymes, dihydroethidium and 4-hydroxy-2-nonenal as indicators for oxidative stress, and anti-inflammatory cytokines (interleukin (IL)-4 and IL-13) and pro-inflammatory cytokines (tumor necrosis factor α (TNF-α) and IL-2) were examined by Western blotting and/or immunohistochemistry. As a result, pretreatment with 30 mg/kg CGA attenuated cognitive impairment and displayed a neuroprotective effect against transient forebrain ischemia (TFI). In Western blotting, the expression levels of SOD2 and IL-4 were increased due to pretreatment with CGA and, furthermore, 4-HNE production and IL-4 expressions were inhibited by CGA pretreatment. Additionally, pretreated CGA enhanced antioxidant enzymes and anti-inflammatory cytokines and, in contrast, attenuated oxidative stress and pro-inflammatory cytokine expression. Based on these results, we suggest that CGA can be a useful neuroprotective material against ischemia-reperfusion injury due to its antioxidant and anti-inflammatory efficacies.
Collapse
|
43
|
Ji Y, Xie Y, Wang T, Cao D, Li J, Han J, Ji G, Zhao S, Kang Z. Four patients with infarction in key areas of the Papez circuit, with anterograde amnesia as the main manifestation. J Int Med Res 2020; 48:300060520939369. [PMID: 32686968 PMCID: PMC7372620 DOI: 10.1177/0300060520939369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The Papez circuit is an important brain structure that is closely associated with
learning and memory. In this report, we present four patients with anterograde amnesia as
the main manifestation induced by Papez circuit infarction. In addition, we review the
distribution of the responsible arteries in key and rare regions to investigate the
pathogenesis of these infarctions.
Collapse
Affiliation(s)
- Ye Ji
- Department of Neurological Function Examination, Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Yanan Xie
- Department of Cardiovascular Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tingting Wang
- Department of Neurology, Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Duanhua Cao
- Department of Neurology, Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Jin Li
- Department of Neurological Function Examination, Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Jingzhe Han
- Department of Neurology, Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Guang Ji
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Song Zhao
- Department of MRI, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhilei Kang
- Department of MRI, Harrison International Peace Hospital, Hengshui, Hebei, China
| |
Collapse
|
44
|
Johnson AC, Miller JE, Cipolla MJ. Memory impairment in spontaneously hypertensive rats is associated with hippocampal hypoperfusion and hippocampal vascular dysfunction. J Cereb Blood Flow Metab 2020; 40:845-859. [PMID: 31088235 PMCID: PMC7168795 DOI: 10.1177/0271678x19848510] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We investigated the effect of chronic hypertension on hippocampal arterioles (HippAs) and hippocampal perfusion as underlying mechanisms of memory impairment, and how large artery stiffness relates to HippA remodeling. Using male spontaneously hypertensive rats (SHR) and normotensive Wistar rats (n = 12/group), long-term (LTM) and spatial memory were tested using object recognition and spontaneous alternation tasks. Hippocampal blood flow was measured via hydrogen clearance basally and during hypercapnia. Reactivity of isolated and pressurized HippAs to pressure and pharmacological activators and inhibitors was investigated. To determine large artery stiffness, distensibility and elastin content were measured in thoracic aorta. SHR had impaired LTM and spatial memory associated with decreased basal blood flow (68 ± 12 mL/100 g/min) vs. Wistar (111 ± 28 mL/100 g/min, p < 0.01) that increased during hypercapnia similarly between groups. Compared to Wistar, HippAs from SHR had increased tone at 60 mmHg (58 ± 9% vs. 37 ± 7%, p < 0.01), and decreased reactivity to small- and intermediate-conductance calcium-activated potassium (SK/IK) channel activation. HippAs in both groups were unaffected by NOS inhibition. Decreased elastin content correlated with increased stiffness in aorta of SHR that was associated with increased stiffness and hypertrophic remodeling of HippAs. Hippocampal vascular dysfunction during hypertension could potentiate memory deficits and may provide a therapeutic target to limit vascular cognitive impairment.
Collapse
Affiliation(s)
- Abbie C Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Justin E Miller
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Marilyn J Cipolla
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA.,Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT, USA
| |
Collapse
|
45
|
Protective Effect of Dimethyl Fumarate on Memory Impairment After Cerebral Ischemia-Reperfusion Injury in Rats. Jundishapur J Nat Pharm Prod 2019. [DOI: 10.5812/jjnpp.69310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
46
|
Heat shock protein 70 increases cell proliferation, neuroblast differentiation, and the phosphorylation of CREB in the hippocampus. Lab Anim Res 2019; 35:21. [PMID: 32257909 PMCID: PMC7081702 DOI: 10.1186/s42826-019-0020-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/09/2019] [Indexed: 12/22/2022] Open
Abstract
In the present study, we investigated the effects of heat shock protein 70 (HSP70) on novel object recognition, cell proliferation, and neuroblast differentiation in the hippocampus. To facilitate penetration into the blood–brain barrier and neuronal plasma membrane, we created a Tat-HSP70 fusion protein. Eight-week-old mice received intraperitoneal injections of vehicle (10% glycerol), control-HSP70, or Tat-HSP70 protein once a day for 21 days. To elucidate the delivery efficiency of HSP70 into the hippocampus, western blot analysis for polyhistidine was conducted. Polyhistidine protein levels were significantly increased in control-HSP70- and Tat-HSP70-treated groups compared to the control or vehicle-treated group. However, polyhistidine protein levels were significantly higher in the Tat-HSP70-treated group compared to that in the control-HSP70-treated group. In addition, immunohistochemical study for HSP70 showed direct evidences for induction of HSP70 immunoreactivity in the control-HSP70- and Tat-HSP70-treated groups. Administration of Tat-HSP70 increased the novel object recognition memory compared to untreated mice or mice treated with the vehicle. In addition, the administration of Tat-HSP70 significantly increased the populations of proliferating cells and differentiated neuroblasts in the dentate gyrus compared to those in the control or vehicle-treated group based on the Ki67 and doublecortin (DCX) immunostaining. Furthermore, the phosphorylation of cAMP response element-binding protein (pCREB) was significantly enhanced in the dentate gyrus of the Tat-HSP70-treated group compared to that in the control or vehicle-treated group. Western blot study also demonstrated the increases of DCX and pCREB protein levels in the Tat-HSP70-treated group compared to that in the control or vehicle-treated group. In contrast, administration of control-HSP70 moderately increased the novel object recognition memory, cell proliferation, and neuroblast differentiation in the dentate gyrus compared to that in the control or vehicle-treated group. These results suggest that Tat-HSP70 promoted hippocampal functions by increasing the pCREB in the hippocampus.
Collapse
|
47
|
El Khashab IH, Abdelsalam RM, Elbrairy AI, Attia AS. Chrysin attenuates global cerebral ischemic reperfusion injury via suppression of oxidative stress, inflammation and apoptosis. Biomed Pharmacother 2019; 112:108619. [DOI: 10.1016/j.biopha.2019.108619] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 01/26/2023] Open
|
48
|
Wang X, Xuan W, Zhu ZY, Li Y, Zhu H, Zhu L, Fu DY, Yang LQ, Li PY, Yu WF. The evolving role of neuro-immune interaction in brain repair after cerebral ischemic stroke. CNS Neurosci Ther 2018; 24:1100-1114. [PMID: 30350341 DOI: 10.1111/cns.13077] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 09/23/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke is the world's leading cause of disability with limited brain repair treatments which effectively improve long-term neurological deficits. The neuroinflammatory responses persist into the late repair phase of stroke and participate in all brain repair elements, including neurogenesis, angiogenesis, synaptogenesis, remyelination and axonal sprouting, shedding new light on post-stroke brain recovery. Resident brain glial cells, such as astrocytes not only contribute to neuroinflammation after stroke, but also secrete a wide range of trophic factors that can promote post-stroke brain repair. Alternatively, activated microglia, monocytes, and neutrophils in the innate immune system, traditionally considered as major damaging factors after stroke, have been suggested to be extensively involved in brain repair after stroke. The adaptive immune system may also have its bright side during the late regenerative phase, affecting the immune suppressive regulatory T cells and B cells. This review summarizes the recent findings in the evolving role of neuroinflammation in multiple post-stroke brain repair mechanisms and poses unanswered questions that may generate new directions for future research and give rise to novel therapeutic targets to improve stroke recovery.
Collapse
Affiliation(s)
- Xin Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Yu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hao Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ling Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dan-Yun Fu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li-Qun Yang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pei-Ying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
49
|
Zhao H, Lu Y, Wang Y, Han X, Zhang Y, Han B, Wang T, Li Y, Wang S. Electroacupunture contributes to recovery of neurological deficits in experimental stroke by activating astrocytes. Restor Neurol Neurosci 2018; 36:301-312. [PMID: 29758953 DOI: 10.3233/rnn-170722] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Neurological deficits is one of the most prevalent clinical manifestation after stroke. The effects of astrocytes activated by electroacupunture (EA) after stroke on the neurological recovery in middle cerebral artery occlusion (MCAO) rats was not clear and definite. OBJECTIVE Our previous study showed that treatment with EA for 7 days contributed to the activation of astrocytes in MCAO rats. The purposes of this study were to 1) confirm the effects of EA for 14 days on activation of astrocytes in MCAO rats, and 2) test the relationships between activation of astrocytes and neurological functional recovery induced by EA in MCAO rats. METHODS All rats were randomly divided into five groups: naïve control group, sham control control group, MCAO, MCAO/EAn, MCAO/EAd (n = 8, for each group). Rats in MCAO/EAn group received EA treatment at acupoints of Neiguan (PC06). MCAO/EAd group received EA stimulus at acupoints of Diji (SP08). The primary indicators were locomotor recovery, histopathology, immunohistochemistry, RT-PCR and Western blot. RESULTS The neurological deficit and histopathological improvements and activation of astrocytes were observed after EA treatment at acupoints PC06. Parametric correlation analyses revealed a cubic correlation relationship between activation of astrocytes and neurological recovery of MCAO rats treated with EA. CONCLUSION EA treatment at the acupoints of Neiguan involved in the regulation of activation of astrocytes, which our data suggested has a cubic correlation relationship with the neurological recovery of MCAO rats.
Collapse
Affiliation(s)
- Haijun Zhao
- Department of Anatomy and Histology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, Shandong, China
| | - Yan Lu
- Department of Experimental Acupuncture Science, College of Acumox and Tuina, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, Shandong, China
| | - Yuan Wang
- Department of Anatomy and Histology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, Shandong, China
| | - Xiaochun Han
- Department of Preventive Medicine, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, Shandong, China
| | - Yanan Zhang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, Shandong, China
| | - Bingbing Han
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, Shandong, China
| | - Tong Wang
- Department of Traditional Chinese Medicine Nursing, College of Nursing Care, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, China
| | - Yan Li
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, Shandong, China.,Department of Pharmaceutical Engineering, School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Changqing District, Jinan, Shandong, China
| | - Shijun Wang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Changqing District, Jinan, Shandong, China
| |
Collapse
|
50
|
Chimeh U, Zimmerman MA, Gilyazova N, Li PA. B355252, A Novel Small Molecule, Confers Neuroprotection Against Cobalt Chloride Toxicity In Mouse Hippocampal Cells Through Altering Mitochondrial Dynamics And Limiting Autophagy Induction. Int J Med Sci 2018; 15:1384-1396. [PMID: 30275767 PMCID: PMC6158673 DOI: 10.7150/ijms.24702] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/12/2018] [Indexed: 12/31/2022] Open
Abstract
Cerebral hypoxia as often occurs in cases of stroke, hemorrhage, or other traumatic brain injuries, is one of the leading causes of death worldwide and a main driver of disabilities in the elderly. Using a chemical mimetic of hypoxia, cobalt chloride (CoCl2), we tested the ability of a novel small molecule, 4-chloro-N-(naphthalen-1-ylmethyl)-5-(3-(piperazin-1-yl)phenoxy)thiophene-2-sulfonamide (B355252), to alleviate CoCl2-induced damage in mouse hippocampal HT22 cells. A dose-dependent decrease in cell viability was observed during CoCl2 treatment along with increases in mitochondrial membrane potential and generation of reactive oxygen species (ROS). B355252 conferred protection against these changes. We further found that mitochondrial dynamics, the balance between mitochondrial fusion and fission, were perturbed by CoCl2 treatment. Mitochondrial fusion, which was assessed by measuring the expression of proteins optic atrophy protein 1 (OPA1) and mitofusin 2 (Mfn2), declined due to CoCl2 exposure, but B355252 addition was able to elevate Mfn2 expression while OPA1 expression was unchanged. Mitochondrial fission, measured by phosphorylated dynamin-related protein 1 (p-DRP1) and fission protein 1 (FIS1) expression, also decreased following CoCl2 exposure, and was stabilized by B355252 addition. Finally, autophagy was assessed by measuring the conversion of cytosolic microtubule-associated protein 1A/1B-light chain three-I (LC3-I) to autophagosome-bound microtubule-associated protein 1A/1B-light chain three-II (LC3-II) and was found to be increased by CoCl2. B355252 addition significantly reduced autophagy induction. Taken together, our results indicate B355252 has therapeutic potential to reduce the damaging effects caused by CoCl2 and should be further evaluated for applications in cerebral ischemia therapy.
Collapse
Affiliation(s)
| | | | | | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC USA
| |
Collapse
|