1
|
Chaudhuri S, Cho M, Stumpff JC, Bice PJ, İş Ö, Ertekin-Taner N, Saykin AJ, Nho K. Cell-specific transcriptional signatures of vascular cells in Alzheimer's disease: perspectives, pathways, and therapeutic directions. Mol Neurodegener 2025; 20:12. [PMID: 39876020 PMCID: PMC11776188 DOI: 10.1186/s13024-025-00798-0] [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: 05/18/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disease that is marked by profound neurovascular dysfunction and significant cell-specific alterations in the brain vasculature. Recent advances in high throughput single-cell transcriptomics technology have enabled the study of the human brain vasculature at an unprecedented depth. Additionally, the understudied niche of cerebrovascular cells, such as endothelial and mural cells, and their subtypes have been scrutinized for understanding cellular and transcriptional heterogeneity in AD. Here, we provide an overview of rich transcriptional signatures derived from recent single-cell and single-nucleus transcriptomic studies of human brain vascular cells and their implications for targeted therapy for AD. We conducted an in-depth literature search using Medline and Covidence to identify pertinent AD studies that utilized single-cell technologies in human post-mortem brain tissue by focusing on understanding the transcriptional differences in cerebrovascular cell types and subtypes in AD and cognitively normal older adults. We also discuss impaired cellular crosstalk between vascular cells and neuroglial units, as well as astrocytes in AD. Additionally, we contextualize the findings from single-cell studies of distinct endothelial cells, smooth muscle cells, fibroblasts, and pericytes in the human AD brain and highlight pathways for potential therapeutic interventions as a concerted multi-omic effort with spatial transcriptomics technology, neuroimaging, and neuropathology. Overall, we provide a detailed account of the vascular cell-specific transcriptional signatures in AD and their crucial cellular crosstalk with the neuroglial unit.
Collapse
Affiliation(s)
- Soumilee Chaudhuri
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Medical Neuroscience Graduate Program, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Minyoung Cho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Julia C Stumpff
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paula J Bice
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Özkan İş
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
2
|
Liu Z, Xia Q, Wang C, Xu J, Tian K, Wang Z, Li L, Li Y, Shang H, Liu Q, Xin T. Biomimetic astrocyte cell membrane-fused nanovesicles for protecting neurovascular units in hypoxic ischemic encephalopathy. J Nanobiotechnology 2024; 22:766. [PMID: 39695691 DOI: 10.1186/s12951-024-03053-8] [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: 09/22/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Hypoxic ischemic encephalopathy (HIE) refers to neonatal hypoxic brain injury caused by severe asphyxia during the perinatal period. With a high incidence rate and poor prognosis, HIE accounts for 2.4% of the global disease burden, imposing a heavy burden on families and society. Current clinical treatment for HIE primarily focuses on symptomatic management and supportive care. Therefore, the developments of effective treatment strategies and new drug formulations are critical for improving the prognosis of HIE patients. In order to protect the compromised neurovascular units after HIE, we prepared membrane-fused nanovesicles for delivering rapamycin and si EDN1 (TRCAM@RAPA@si EDN1). Due to the homotypic targeting feature of membrane-fused nanovesicles, we employed astrocyte membranes as synthetic materials to improve the targeting of astrocytes in brain while reducing the clearance of nanovesicles by circulatory system. Additionally, the surface of cell membrane was modified with CXCR3 receptors, enhancing the homing of nanovesicles to infarcted lesions. Lipid vesicles were modified with TK and RVG29 transmembrane peptides, enabling responsive release of internal drugs and blood-brain barrier penetration. Internally loaded rapamycin could promote protective autophagy in astrocytes, improve cellular oxidative stress, while si EDN1 could reduce the expression level of endothelin gene, thereby reducing secondary damage to neurovascular units.
Collapse
Affiliation(s)
- Zihao Liu
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250021, China
| | - Qian Xia
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chanyue Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jiacan Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Kangqian Tian
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zhihai Wang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Longji Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Yuchen Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Hao Shang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Qian Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China.
| |
Collapse
|
3
|
Liang C, Wei S, Ji Y, Lin J, Jiao W, Li Z, Yan F, Jing X. The role of enteric nervous system and GDNF in depression: Conversation between the brain and the gut. Neurosci Biobehav Rev 2024; 167:105931. [PMID: 39447778 DOI: 10.1016/j.neubiorev.2024.105931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Depression is a debilitating mental disorder that causes a persistent feeling of sadness and loss of interest. Approximately 280 million individuals worldwide suffer from depression by 2023. Despite the heavy medical and social burden imposed by depression, pathophysiology remains incompletely understood. Emerging evidence indicates various bidirectional interplay enable communication between the gut and brain. These interplays provide a link between intestinal and central nervous system as well as feedback from cortical and sensory centers to enteric activities, which also influences physiology and behavior in depression. This review aims to overview the significant role of the enteric nervous system (ENS) in the pathophysiology of depression and gut-brain axis's contribution to depressive disorders. Additionally, we explore the alterations in enteric glia cells (EGCs) and glial cell line-derived neurotrophic factor (GDNF) in depression and their involvement in neuronal support, intestinal homeostasis maintains and immune response activation. Modulating ENS function, EGCs and GDNF level could serve as novel strategies for future antidepressant therapy.
Collapse
Affiliation(s)
- Chuoyi Liang
- School of Nursing, Jinan University, Guangzhou, China
| | - Sijia Wei
- School of Nursing, Jinan University, Guangzhou, China
| | - Yelin Ji
- School of Nursing, Jinan University, Guangzhou, China
| | - Jiayi Lin
- School of Nursing, Jinan University, Guangzhou, China
| | - Wenli Jiao
- School of Nursing, Jinan University, Guangzhou, China
| | - Zhiying Li
- School of Nursing, Jinan University, Guangzhou, China
| | - Fengxia Yan
- School of Nursing, Jinan University, Guangzhou, China.
| | - Xi Jing
- School of Nursing, Jinan University, Guangzhou, China; Guangdong-Hong Kong-Macau Great Bay Area Geoscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
4
|
Hammer MF, Bahramnejad E, Watkins JC, Ronaldson PT. Candesartan restores blood-brain barrier dysfunction, mitigates aberrant gene expression, and extends lifespan in a knockin mouse model of epileptogenesis. Clin Sci (Lond) 2024; 138:1089-1110. [PMID: 39092536 DOI: 10.1042/cs20240771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/04/2024]
Abstract
Blockade of Angiotensin type 1 receptor (AT1R) has potential therapeutic utility in the treatment of numerous detrimental consequences of epileptogenesis, including oxidative stress, neuroinflammation, and blood-brain barrier (BBB) dysfunction. We have recently shown that many of these pathological processes play a critical role in seizure onset and propagation in the Scn8a-N1768D mouse model. Here we investigate the efficacy and potential mechanism(s) of action of candesartan (CND), an FDA-approved angiotensin receptor blocker (ARB) indicated for hypertension, in improving outcomes in this model of pediatric epilepsy. We compared length of lifespan, seizure frequency, and BBB permeability in juvenile (D/D) and adult (D/+) mice treated with CND at times after seizure onset. We performed RNAseq on hippocampal tissue to quantify differences in genome-wide patterns of transcript abundance and inferred beneficial and detrimental effects of canonical pathways identified by enrichment methods in untreated and treated mice. Our results demonstrate that treatment with CND gives rise to increased survival, longer periods of seizure freedom, and diminished BBB permeability. CND treatment also partially reversed or 'normalized' disease-induced genome-wide gene expression profiles associated with inhibition of NF-κB, TNFα, IL-6, and TGF-β signaling in juvenile and adult mice. Pathway analyses reveal that efficacy of CND is due to its known dual mechanism of action as both an AT1R antagonist and a PPARγ agonist. The robust effectiveness of CND across ages, sexes and mouse strains is a positive indication for its translation to humans and its suitability of use for clinical trials in children with SCN8A epilepsy.
Collapse
Affiliation(s)
- Michael F Hammer
- BIO5 Institute, University of Arizona, Tucson, AZ, U.S.A
- Department of Neurology, University of Arizona, Tucson, AZ, U.S.A
| | - Erfan Bahramnejad
- BIO5 Institute, University of Arizona, Tucson, AZ, U.S.A
- Department of Pharmacology, University of Arizona, Tucson, AZ, U.S.A
| | - Joseph C Watkins
- Department of Mathematics, University of Arizona, Tucson, AZ, U.S.A
| | | |
Collapse
|
5
|
Walter AE, Savalia K, Yoon J, Morrison J, Schneider ALC, Diaz-Arrastia R, Sandsmark DK. Change in Enlarged Perivascular Spaces over Time and Associations with Outcomes After Traumatic Brain Injury. Neurotrauma Rep 2024; 5:738-748. [PMID: 39144451 PMCID: PMC11319858 DOI: 10.1089/neur.2024.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024] Open
Abstract
Enlarged perivascular spaces (EPVs) can be seen on magnetic resonance imaging (MRI) scans in various neurological diseases, including traumatic brain injury (TBI). EPVs have been associated with cognitive dysfunction and sleep disturbances; however, their clinical significance remains unclear. The goal of this study was to identify MRI burden of EPVs over time following TBI and to explore their relationship with postinjury outcomes. Individuals with TBI underwent postinjury data collection at Day 1 (blood), 2 weeks (blood, MRI, outcomes), and 6 months (blood, MRI, outcomes). EPV burden was assessed using T1 and FLAIR sequences on representative slices in the centrum semiovale, basal ganglia, and midbrain. Serum blood was assayed to measure concentrations of neurofilament light (NfL) and glial fibrillary acidic protein (GFAP). Thirty-two participants with TBI were included (mean age 36.8 years, 78% male, 50% White). Total EPVs count did not significantly change from 2 weeks (23.5 [95% confidence interval or CI = 22.0-32.0]) to 6 months (26.0 [95% CI = 22.0-30.0], p = 0.16). For self-reported measures of sleep, there were no significant associations between EPVs count and Insomnia Severity Index (2 weeks: β = -0.004; 95% CI = -0.094, 0.086; 6 months: β = 0.002; 95% CI = -0.122, 0.125) or the subset of sleep questions on the Rivermead Post-Concussion Symptoms Questionnaire (2 weeks: β = -0.005; 95% CI = -0.049, 0.039; 6 months: β = -0.019; 95% CI = -0.079, 0.042). Functional outcome, determined by 6 months incomplete recovery (Glasgow Outcome Scale-Extended [GOS-E < 8]) versus complete recovery (GOS-E = 8), was significantly associated with a higher number of EPVs at 2 weeks (odds ratio = 0.94, 95% CI = 0.88-0.99). Spearman correlations showed no significant relationship between EPVs count and GFAP or NfL. This study used commonly acquired MRI sequences to quantify EPVs and investigated their utility as a potential imaging biomarker in TBI. Given the minimal change in EPVs over time, this period may not be long enough for potential recovery or may indicate that EPVs are structural findings that do not significantly change over time.
Collapse
Affiliation(s)
- Alexa E. Walter
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Krupa Savalia
- Departments of Neurology and Neurological Surgery, University of California Davis, Davis, California, USA
| | - Jason Yoon
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Justin Morrison
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrea L. C. Schneider
- Departments of Neurology and Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Danielle K. Sandsmark
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Ding Y, Peng YY, Li S, Tang C, Gao J, Wang HY, Long ZY, Lu XM, Wang YT. Single-Cell Sequencing Technology and Its Application in the Study of Central Nervous System Diseases. Cell Biochem Biophys 2024; 82:329-342. [PMID: 38133792 DOI: 10.1007/s12013-023-01207-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
The mammalian central nervous system consists of a large number of cells, which contain not only different types of neurons, but also a large number of glial cells, such as astrocytes, oligodendrocytes, and microglia. These cells are capable of performing highly refined electrophysiological activities and providing the brain with functions such as nutritional support, information transmission and pathogen defense. The diversity of cell types and individual differences between cells have brought inspiration to the study of the mechanism of central nervous system diseases. In order to explore the role of different cells, a new technology, single-cell sequencing technology has emerged to perform specific analysis of high-throughput cell populations, and has been continuously developed. Single-cell sequencing technology can accurately analyze single-cell expression in mixed-cell populations and collect cells from different spatial locations, time stages and types. By using single-cell sequencing technology to compare gene expression profiles of normal and diseased cells, it is possible to discover cell subsets associated with specific diseases and their associated genes. Therefore, scientists can understand the development process, related functions and disease state of the nervous system from an unprecedented depth. In conclusion, single-cell sequencing technology provides a powerful technology for the discovery of novel therapeutic targets for central nervous system diseases.
Collapse
Affiliation(s)
- Yang Ding
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yu-Yuan Peng
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Sen Li
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Can Tang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jie Gao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zai-Yun Long
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
| | - Yong-Tang Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
7
|
Huber RE, Babbitt C, Peyton SR. Heterogeneity of brain extracellular matrix and astrocyte activation. J Neurosci Res 2024; 102:e25356. [PMID: 38773875 DOI: 10.1002/jnr.25356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/01/2024] [Accepted: 05/05/2024] [Indexed: 05/24/2024]
Abstract
From the blood brain barrier to the synaptic space, astrocytes provide structural, metabolic, ionic, and extracellular matrix (ECM) support across the brain. Astrocytes include a vast array of subtypes, their phenotypes and functions varying both regionally and temporally. Astrocytes' metabolic and regulatory functions poise them to be quick and sensitive responders to injury and disease in the brain as revealed by single cell sequencing. Far less is known about the influence of the local healthy and aging microenvironments on these astrocyte activation states. In this forward-looking review, we describe the known relationship between astrocytes and their local microenvironment, the remodeling of the microenvironment during disease and injury, and postulate how they may drive astrocyte activation. We suggest technology development to better understand the dynamic diversity of astrocyte activation states, and how basal and activation states depend on the ECM microenvironment. A deeper understanding of astrocyte response to stimuli in ECM-specific contexts (brain region, age, and sex of individual), paves the way to revolutionize how the field considers astrocyte-ECM interactions in brain injury and disease and opens routes to return astrocytes to a healthy quiescent state.
Collapse
Affiliation(s)
- Rebecca E Huber
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Courtney Babbitt
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
8
|
Czyżewski W, Mazurek M, Sakwa L, Szymoniuk M, Pham J, Pasierb B, Litak J, Czyżewska E, Turek M, Piotrowski B, Torres K, Rola R. Astroglial Cells: Emerging Therapeutic Targets in the Management of Traumatic Brain Injury. Cells 2024; 13:148. [PMID: 38247839 PMCID: PMC10813911 DOI: 10.3390/cells13020148] [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: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Traumatic Brain Injury (TBI) represents a significant health concern, necessitating advanced therapeutic interventions. This detailed review explores the critical roles of astrocytes, key cellular constituents of the central nervous system (CNS), in both the pathophysiology and possible rehabilitation of TBI. Following injury, astrocytes exhibit reactive transformations, differentiating into pro-inflammatory (A1) and neuroprotective (A2) phenotypes. This paper elucidates the interactions of astrocytes with neurons, their role in neuroinflammation, and the potential for their therapeutic exploitation. Emphasized strategies encompass the utilization of endocannabinoid and calcium signaling pathways, hormone-based treatments like 17β-estradiol, biological therapies employing anti-HBGB1 monoclonal antibodies, gene therapy targeting Connexin 43, and the innovative technique of astrocyte transplantation as a means to repair damaged neural tissues.
Collapse
Affiliation(s)
- Wojciech Czyżewski
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland;
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| | - Marek Mazurek
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| | - Leon Sakwa
- Student Scientific Society, Kazimierz Pulaski University of Radom, 26-600 Radom, Poland;
| | - Michał Szymoniuk
- Student Scientific Association, Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Jennifer Pham
- Student Scientific Society, Medical University of Lublin, 20-954 Lublin, Poland; (J.P.); (M.T.)
| | - Barbara Pasierb
- Department of Dermatology, Radom Specialist Hospital, 26-600 Radom, Poland;
| | - Jakub Litak
- Department of Clinical Immunology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Ewa Czyżewska
- Department of Otolaryngology, Mazovian Specialist Hospital, 26-617 Radom, Poland;
| | - Michał Turek
- Student Scientific Society, Medical University of Lublin, 20-954 Lublin, Poland; (J.P.); (M.T.)
| | - Bartłomiej Piotrowski
- Institute of Automatic Control and Robotics, Warsaw University of Technology, 00-661 Warsaw, Poland;
| | - Kamil Torres
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Radosław Rola
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| |
Collapse
|
9
|
Blaber AP, Sadeghian F, Naz Divsalar D, Scarisbrick IA. Elevated biomarkers of neural injury in older adults following head-down bed rest: links to cardio-postural deconditioning with spaceflight and aging. Front Hum Neurosci 2023; 17:1208273. [PMID: 37822710 PMCID: PMC10562592 DOI: 10.3389/fnhum.2023.1208273] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction Prolonged physical inactivity with bed rest or spaceflight is associated with cardiovascular and neuromuscular deconditioning; however, its impact on neural integrity of cardio-postural reflexes and possible mitigation with exercise has not been examined. We assessed the association between the physiological deconditioning of bed rest immobilization with neural injury markers and the effects of 60-75 min of daily exercise. Methods Data were collected as part of a randomized clinical trial (clinicaltrials.gov identifier: NCT04964999) at the McGill University Medical Centre. Twenty-two 55- to 65-year-old healthy volunteers gave informed consent and took part. Within sex, participants were randomly assigned to exercise (60- to 75-min daily) or control (inactive) groups and spent 14 days in continuous 6° head-down tilt. Neural injury [neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), total tau (t-Tau), myelin basic protein (MBP), brain-derived neurotrophic factor (BDNF), ubiquitin carboxy-terminal hydrolase L1 (UCH-L1)], as well as interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and insulin-like growth factor 1 (IGF-1) biomarkers were measured before, during, and after bed rest. The false discovery rate with Huber M-estimation was used to correlate changes in biomarkers with cardiovascular and muscular function changes over bed rest. Results Bed rest elevated NfL, GFAP, TNF-α, and IL-6 in all participants and reduced IGF-1 in females only. With standing, changes in heart rate, blood pressure, and lower limb muscle motoneuron activity correlated with changes in TNF-α and BDNF. Baroreflex control, leg muscle maximal voluntary contraction, and postural sway are correlated with GFAP and NfL. Exercise participants had fewer interactions than control participants, but significant correlations still existed, with both groups exhibiting similar reductions in orthostatic tolerance. Discussion An hour of daily exercise in older persons otherwise immobilized for 2 weeks did not abate bed rest-induced increases in serum signatures of neural injury or pro-inflammatory markers. Exercise reduced the number of physiological interactions of biomarkers, but significant cardio-postural correlations remained with no protection against post-bed rest orthostatic intolerance. The identification of associations of inflammatory and neural injury biomarkers with changes in cardio-postural physiology and exercise points to biotherapeutic opportunities and improved exercise interventions for astronauts and individuals in bed rest. Clinical trial registration https://www.clinicaltrials.gov/search?cond=NCT04964999, identifier: NCT04964999.
Collapse
Affiliation(s)
- Andrew P. Blaber
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Farshid Sadeghian
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Donya Naz Divsalar
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Isobel A. Scarisbrick
- Department of Physical Medicine and Rehabilitation, Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
10
|
Zeng CW, Tsai HJ. The Promising Role of a Zebrafish Model Employed in Neural Regeneration Following a Spinal Cord Injury. Int J Mol Sci 2023; 24:13938. [PMID: 37762240 PMCID: PMC10530783 DOI: 10.3390/ijms241813938] [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: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating event that results in a wide range of physical impairments and disabilities. Despite the advances in our understanding of the biological response to injured tissue, no effective treatments are available for SCIs at present. Some studies have addressed this issue by exploring the potential of cell transplantation therapy. However, because of the abnormal microenvironment in injured tissue, the survival rate of transplanted cells is often low, thus limiting the efficacy of such treatments. Many studies have attempted to overcome these obstacles using a variety of cell types and animal models. Recent studies have shown the utility of zebrafish as a model of neural regeneration following SCIs, including the proliferation and migration of various cell types and the involvement of various progenitor cells. In this review, we discuss some of the current challenges in SCI research, including the accurate identification of cell types involved in neural regeneration, the adverse microenvironment created by SCIs, attenuated immune responses that inhibit nerve regeneration, and glial scar formation that prevents axonal regeneration. More in-depth studies are needed to fully understand the neural regeneration mechanisms, proteins, and signaling pathways involved in the complex interactions between the SCI microenvironment and transplanted cells in non-mammals, particularly in the zebrafish model, which could, in turn, lead to new therapeutic approaches to treat SCIs in humans and other mammals.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Huai-Jen Tsai
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| |
Collapse
|
11
|
Stanca S, Rossetti M, Bongioanni P. Astrocytes as Neuroimmunocytes in Alzheimer's Disease: A Biochemical Tool in the Neuron-Glia Crosstalk along the Pathogenetic Pathways. Int J Mol Sci 2023; 24:13880. [PMID: 37762184 PMCID: PMC10531177 DOI: 10.3390/ijms241813880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
This work aimed at assessing Alzheimer's disease (AD) pathogenesis through the investigation of the astrocytic role to transduce the load of amyloid-beta (Aβ) into neuronal death. The backbone of this review is focused on the deepening of the molecular pathways eliciting the activation of astrocytes crucial phenomena in the understanding of AD as an autoimmune pathology. The complex relations among astrocytes, Aβ and tau, together with the role played by the tripartite synapsis are discussed. A review of studies published from 1979 to 2023 on Scopus, PubMed and Google Scholar databases was conducted. The selected papers focused not only on the morphological and metabolic characteristics of astrocytes, but also on the latest notions about their multifunctional involvement in AD pathogenesis. Astrocytes participate in crucial pathways, including pruning and sprouting, by which the AD neurodegeneration evolves from an aggregopathy to neuroinflammation, loss of synapses and neuronal death. A1 astrocytes stimulate the production of pro-inflammatory molecules which have been correlated with the progression of AD cognitive impairment. Further research is needed to "hold back" the A1 polarization and, thus, to slow the worsening of the disease. AD clinical expression is the result of dysfunctional neuronal interactions, but this is only the end of a process involving a plurality of protagonists. One of these is the astrocyte, whose importance this work intends to put under the spotlight in the AD scenario, reflecting the multifaceted nature of this disease in the functional versatility of this glial population.
Collapse
Affiliation(s)
- Stefano Stanca
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy
- NeuroCare Onlus, 56100 Pisa, Italy
| | - Martina Rossetti
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy
- NeuroCare Onlus, 56100 Pisa, Italy
| | - Paolo Bongioanni
- NeuroCare Onlus, 56100 Pisa, Italy
- Medical Specialties Department, Azienda Ospedaliero-Universitaria Pisana, 56100 Pisa, Italy
| |
Collapse
|
12
|
Sfera A, Rahman L, Zapata-Martín Del Campo CM, Kozlakidis Z. Long COVID as a Tauopathy: Of "Brain Fog" and "Fusogen Storms". Int J Mol Sci 2023; 24:12648. [PMID: 37628830 PMCID: PMC10454863 DOI: 10.3390/ijms241612648] [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: 07/13/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Long COVID, also called post-acute sequelae of SARS-CoV-2, is characterized by a multitude of lingering symptoms, including impaired cognition, that can last for many months. This symptom, often called "brain fog", affects the life quality of numerous individuals, increasing medical complications as well as healthcare expenditures. The etiopathogenesis of SARS-CoV-2-induced cognitive deficit is unclear, but the most likely cause is chronic inflammation maintained by a viral remnant thriving in select body reservoirs. These viral sanctuaries are likely comprised of fused, senescent cells, including microglia and astrocytes, that the pathogen can convert into neurotoxic phenotypes. Moreover, as the enteric nervous system contains neurons and glia, the virus likely lingers in the gastrointestinal tract as well, accounting for the intestinal symptoms of long COVID. Fusogens are proteins that can overcome the repulsive forces between cell membranes, allowing the virus to coalesce with host cells and enter the cytoplasm. In the intracellular compartment, the pathogen hijacks the actin cytoskeleton, fusing host cells with each other and engendering pathological syncytia. Cell-cell fusion enables the virus to infect the healthy neighboring cells. We surmise that syncytia formation drives cognitive impairment by facilitating the "seeding" of hyperphosphorylated Tau, documented in COVID-19. In our previous work, we hypothesized that the SARS-CoV-2 virus induces premature endothelial senescence, increasing the permeability of the intestinal and blood-brain barrier. This enables the migration of gastrointestinal tract microbes and/or their components into the host circulation, eventually reaching the brain where they may induce cognitive dysfunction. For example, translocated lipopolysaccharides or microbial DNA can induce Tau hyperphosphorylation, likely accounting for memory problems. In this perspective article, we examine the pathogenetic mechanisms and potential biomarkers of long COVID, including microbial cell-free DNA, interleukin 22, and phosphorylated Tau, as well as the beneficial effect of transcutaneous vagal nerve stimulation.
Collapse
Affiliation(s)
- Adonis Sfera
- Paton State Hospital, 3102 Highland Ave, Patton, CA 92369, USA
- School of Behavioral Health, Loma Linda University, 11139 Anderson St., Loma Linda, CA 92350, USA
- Department of Psychiatry, University of California, Riverside 900 University Ave, Riverside, CA 92521, USA
| | - Leah Rahman
- Department of Neuroscience, University of Oregon, 222 Huestis Hall, Eugene, OR 97401, USA
| | | | - Zisis Kozlakidis
- International Agency for Research on Cancer, World Health Organization, 69000 Lyon, France
| |
Collapse
|
13
|
Guebel DV. Human hippocampal astrocytes: Computational dissection of their transcriptome, sexual differences and exosomes across ageing and mild-cognitive impairment. Eur J Neurosci 2023; 58:2677-2707. [PMID: 37427765 DOI: 10.1111/ejn.16081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/20/2023] [Accepted: 06/16/2023] [Indexed: 07/11/2023]
Abstract
The role of astrocytes in Alzheimer's disease is often disregarded. Hence, characterization of astrocytes along their early evolution toward Alzheimer would be greatly beneficial. However, due to their exquisite responsiveness, in vivo studies are difficult. So public microarray data of hippocampal homogenates from (healthy) young, (healthy) elder and elder with mild cognitive impairment (MCI) were subjected to re-analysis by a multi-step computational pipeline. Ontologies and pathway analyses were compared after determining the differential genes that, belonging to astrocytes, have splice forms. Likewise, the subset of molecules exportable to exosomes was also determined. The results showed that astrocyte's phenotypes changed significantly. While already 'activated' astrocytes were found in the younger group, major changes occurred during ageing (increased vascular remodelling and response to mechanical stimulus, diminished long-term potentiation and increased long-term depression). MCI's astrocytes showed some 'rejuvenated' features, but their sensitivity to shear stress was markedly lost. Importantly, most of the changes showed to be sex biassed. Men's astrocytes are enriched in a type 'endfeet-astrocytome', whereas women's astrocytes appear close to the 'scar-forming' type (prone to endothelial dysfunction, hypercholesterolemia, loss of glutamatergic synapses, Ca+2 dysregulation, hypoxia, oxidative stress and 'pro-coagulant' phenotype). In conclusion, the computational dissection of the networks based on the hippocampal gene isoforms provides a relevant proxy to in vivo astrocytes, also revealing the occurrence of sexual differences. Analyses of the astrocytic exosomes did not provide an acceptable approximation to the overall functioning of astrocytes in the hippocampus, probably due to the selective cellular mechanisms which charge the cargo molecules.
Collapse
|
14
|
Sun T, Feng M, Manyande A, Xiang H, Xiong J, He Z. Regulation of mild cognitive impairment associated with liver disease by humoral factors derived from the gastrointestinal tract and MRI research progress: a literature review. Front Neurosci 2023; 17:1206417. [PMID: 37397455 PMCID: PMC10312011 DOI: 10.3389/fnins.2023.1206417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Patients with liver disease are prone to various cognitive impairments. It is undeniable that cognitive impairment is often regulated by both the nervous system and the immune system. In this review our research focused on the regulation of mild cognitive impairment associated with liver disease by humoral factors derived from the gastrointestinal tract, and revealed that its mechanisms may be involved with hyperammonemia, neuroinflammation, brain energy and neurotransmitter metabolic disorders, and liver-derived factors. In addition, we share the emerging research progress in magnetic resonance imaging techniques of the brain during mild cognitive impairment associated with liver disease, in order to provide ideas for the prevention and treatment of mild cognitive impairment in liver disease.
Collapse
Affiliation(s)
- Tianning Sun
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Maohui Feng
- Department of Gastrointestinal Surgery, Wuhan Peritoneal Cancer Clinical Medical Research Center, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, United Kingdom
| | - Hongbing Xiang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Xiong
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhigang He
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
15
|
Zaninello M, Bean C. Highly Specialized Mechanisms for Mitochondrial Transport in Neurons: From Intracellular Mobility to Intercellular Transfer of Mitochondria. Biomolecules 2023; 13:938. [PMID: 37371518 DOI: 10.3390/biom13060938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/26/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
The highly specialized structure and function of neurons depend on a sophisticated organization of the cytoskeleton, which supports a similarly sophisticated system to traffic organelles and cargo vesicles. Mitochondria sustain crucial functions by providing energy and buffering calcium where it is needed. Accordingly, the distribution of mitochondria is not even in neurons and is regulated by a dynamic balance between active transport and stable docking events. This system is finely tuned to respond to changes in environmental conditions and neuronal activity. In this review, we summarize the mechanisms by which mitochondria are selectively transported in different compartments, taking into account the structure of the cytoskeleton, the molecular motors and the metabolism of neurons. Remarkably, the motor proteins driving the mitochondrial transport in axons have been shown to also mediate their transfer between cells. This so-named intercellular transport of mitochondria is opening new exciting perspectives in the treatment of multiple diseases.
Collapse
Affiliation(s)
- Marta Zaninello
- Institute for Genetics, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Camilla Bean
- Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
16
|
Shen X, Li M, Shao K, Li Y, Ge Z. Post-ischemic inflammatory response in the brain: Targeting immune cell in ischemic stroke therapy. Front Mol Neurosci 2023; 16:1076016. [PMID: 37078089 PMCID: PMC10106693 DOI: 10.3389/fnmol.2023.1076016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
An ischemic stroke occurs when the blood supply is obstructed to the vascular basin, causing the death of nerve cells and forming the ischemic core. Subsequently, the brain enters the stage of reconstruction and repair. The whole process includes cellular brain damage, inflammatory reaction, blood–brain barrier destruction, and nerve repair. During this process, the proportion and function of neurons, immune cells, glial cells, endothelial cells, and other cells change. Identifying potential differences in gene expression between cell types or heterogeneity between cells of the same type helps to understand the cellular changes that occur in the brain and the context of disease. The recent emergence of single-cell sequencing technology has promoted the exploration of single-cell diversity and the elucidation of the molecular mechanism of ischemic stroke, thus providing new ideas and directions for the diagnosis and clinical treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xueyang Shen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Mingming Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Gansu Provincial Neurology Clinical Medical Research Center, The Second Hospital of Lanzhou University, Lanzhou, China
- Expert Workstation of Academician Wang Longde, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Kangmei Shao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Yongnan Li,
| | - Zhaoming Ge
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Gansu Provincial Neurology Clinical Medical Research Center, The Second Hospital of Lanzhou University, Lanzhou, China
- Expert Workstation of Academician Wang Longde, The Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Zhaoming Ge,
| |
Collapse
|
17
|
Kalinichenko SG, Pushchin II, Matveeva NY. Neurotoxic and cytoprotective mechanisms in the ischemic neocortex. J Chem Neuroanat 2023; 128:102230. [PMID: 36603664 DOI: 10.1016/j.jchemneu.2022.102230] [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: 09/19/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Neuronal damage in ischemic stroke occurs due to permanent imbalance between the metabolic needs of the brain and the ability of the blood-vascular system to maintain glucose delivery and adequate gas exchange. Oxidative stress and excitotoxicity trigger complex processes of neuroinflammation, necrosis, and apoptosis of both neurons and glial cells. This review summarizes data on the structural and chemical changes in the neocortex and main cytoprotective effects induced by focal ischemic stroke. We focus on the expression of neurotrophins (NT) and molecular and cellular changes in neurovascular units in ischemic brain. We also discuss how these factors affect the apoptosis of cortical cells. Ischemic damage involves close interaction of a wide range of signaling molecules, each acting as an efficient marker of cell state in both the ischemic core and penumbra. NTs play the main regulatory role in brain tissue recovery after ischemic injury. Heterogeneous distribution of the BDNF, NT-3, and GDNF immunoreactivity is concordant with the selective response of different types of cortical neurons and glia to ischemic injury and allows mapping the position of viable neurons. Astrocytes are the central link in neurovascular coupling in ischemic brain by providing other cells with a wide range of vasotropic factors. The NT expression coincides with the distribution of reactive astrocytes, marking the boundaries of the penumbra. The development of ischemic stroke is accompanied by a dramatic change in the distribution of GDNF reactivity. In early ischemic period, it is mainly observed in cortical neurons, while in late one, the bulk of GDNF-positive cells are various types of glia, in particular, astrocytes. The proportion of GDNF-positive astrocytes increases gradually throughout the ischemic period. Some factors that exert cytoprotective effects in early ischemic period may display neurotoxic and pro-apoptotic effects later on. The number of apoptotic cells in the ischemic brain tissue correlates with the BDNF levels, corroborating its protective effects. Cytoprotection and neuroplasticity are two lines of brain protection and recovery after ischemic stroke. NTs can be considered an important link in these processes. To develop efficient pharmacological therapy for ischemic brain injury, we have to deepen our understanding of neurochemical adaptation of brain tissue to acute stroke.
Collapse
Affiliation(s)
- Sergei G Kalinichenko
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| | - Igor I Pushchin
- Laboratory of Physiology, A.V. Zhirmusky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.
| | - Natalya Yu Matveeva
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| |
Collapse
|
18
|
Henao‐Restrepo J, López‐Murillo C, Valderrama‐Carmona P, Orozco‐Santa N, Gomez J, Gutiérrez‐Vargas J, Moraga R, Toledo J, Littau JL, Härtel S, Arboleda‐Velásquez JF, Sepulveda‐Falla D, Lopera F, Cardona‐Gómez GP, Villegas A, Posada‐Duque R. Gliovascular alterations in sporadic and familial Alzheimer's disease: APOE3 Christchurch homozygote glioprotection. Brain Pathol 2023; 33:e13119. [PMID: 36130084 PMCID: PMC10041169 DOI: 10.1111/bpa.13119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
In response to brain insults, astrocytes become reactive, promoting protection and tissue repair. However, astroglial reactivity is typical of brain pathologies, including Alzheimer's disease (AD). Considering the heterogeneity of the reactive response, the role of astrocytes in the course of different forms of AD has been underestimated. Colombia has the largest human group known to have familial AD (FAD). This group carries the autosomal dominant and fully penetrant mutation E280A in PSEN1, which causes early-onset AD. Recently, our group identified an E280A carrier who did not develop FAD. The individual was homozygous for the Christchurch mutation R136S in APOE3 (APOEch). Remarkably, APOE is the main genetic risk factor for developing sporadic AD (SAD) and most of cerebral ApoE is produced by astroglia. Here, we characterized astrocyte properties related to reactivity, glutamate homeostasis, and structural integrity of the gliovascular unit (GVU), as factors that could underlie the pathogenesis or protection of AD. Specifically, through histological and 3D microscopy analyses of postmortem samples, we briefly describe the histopathology and cytoarchitecture of the frontal cortex of SAD, FAD, and APOEch, and demonstrate that, while astrodegeneration and vascular deterioration are prominent in SAD, FAD is characterized by hyperreactive-like glia, and APOEch displays the mildest astrocytic and vascular alterations despite having the highest burden of Aβ. Notably, astroglial, gliovascular, and vascular disturbances, as well as brain cell death, correlate with the specific astrocytic phenotypes identified in each condition. This study provides new insights into the potential relevance of the gliovasculature in the development and protection of AD. To our knowledge, this is the first study assessing the components of the GVU in human samples of SAD, FAD, and APOEch.
Collapse
Affiliation(s)
- Julián Henao‐Restrepo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Carolina López‐Murillo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Pablo Valderrama‐Carmona
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Natalia Orozco‐Santa
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Johana Gomez
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Johanna Gutiérrez‐Vargas
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Health Sciences FacultyRemington University CorporationMedellínColombia
| | - Renato Moraga
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jorge Toledo
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jessica Lisa Littau
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Steffen Härtel
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Joseph F. Arboleda‐Velásquez
- Schepens Eye Research Institute of Mass Eye and Ear, Department of OphthalmologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Diego Sepulveda‐Falla
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Gloria Patricia Cardona‐Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Andrés Villegas
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Rafael Posada‐Duque
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| |
Collapse
|
19
|
Zargari M, Meyer LJ, Riess ML, Li Z, Barajas MB. P188 Therapy in In Vitro Models of Traumatic Brain Injury. Int J Mol Sci 2023; 24:3334. [PMID: 36834743 PMCID: PMC9961452 DOI: 10.3390/ijms24043334] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Traumatic brain injury (TBI) is a significant cause of morbidity and mortality worldwide. Varied mechanisms of injury contribute to the heterogeneity of this patient population as demonstrated by the multiple published grading scales and diverse required criteria leading to diagnoses from mild to severe. TBI pathophysiology is classically separated into a primary injury that is characterized by local tissue destruction as a result of the initial blow, followed by a secondary phase of injury constituted by a score of incompletely understood cellular processes including reperfusion injury, disruption to the blood-brain barrier, excitotoxicity, and metabolic dysregulation. There are currently no effective pharmacological treatments in the wide-spread use for TBI, in large part due to challenges associated with the development of clinically representative in vitro and in vivo models. Poloxamer 188 (P188), a Food and Drug Administration-approved amphiphilic triblock copolymer embeds itself into the plasma membrane of damaged cells. P188 has been shown to have neuroprotective properties on various cell types. The objective of this review is to provide a summary of the current literature on in vitro models of TBI treated with P188.
Collapse
Affiliation(s)
- Michael Zargari
- Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | - Matthias L. Riess
- TVHS VA Medical Center, Anesthesiology, Nashville, TN 37212, USA
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Zhu Li
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew B. Barajas
- TVHS VA Medical Center, Anesthesiology, Nashville, TN 37212, USA
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
20
|
Liang J, Min LQ, Zhu XY, Ma TT, Li Y, Zhang MQ, Zhao L. Fingolimod protects against neurovascular unit injury in a rat model of focal cerebral ischemia/reperfusion injury. Neural Regen Res 2023; 18:869-874. [DOI: 10.4103/1673-5374.353500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
21
|
The Reactive Astrocytes After Surgical Brain Injury Potentiates the Migration, Invasion, and Angiogenesis of C6 Glioma. World Neurosurg 2022; 168:e595-e606. [DOI: 10.1016/j.wneu.2022.10.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
|
22
|
Khan H, Kaur Grewal A, Gurjeet Singh T. Mitochondrial dynamics related neurovascular approaches in cerebral ischemic injury. Mitochondrion 2022; 66:54-66. [DOI: 10.1016/j.mito.2022.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/14/2022] [Accepted: 08/02/2022] [Indexed: 12/30/2022]
|
23
|
Kit O, Frantsiyants E, Neskubina I, Shikhlyarova A, Kaplieva I. Mitochondrial therapy: a vision of the outlooks for treatment of main twenty-first-century diseases. CARDIOMETRY 2022. [DOI: 10.18137/cardiometry.2022.22.1827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are dynamic organelles which constantly change their shape, size, and location within the cells. Mitochondrial dynamics is associated with mesenchymal metabolism or epithelial-mesenchymal transition to regulate the stem cell differentiation, proliferation, migration, and apoptosis. The transfer of mitochondria from one cell to another is necessary to improve and maintain homeostasis in an organism. Mitochondrial transplantation is a therapeutic approach that involves an introduction of healthy mitochondria into damaged organs. Recent evidence data have shown that the physiological properties of healthy mitochondria provide their ability to replace damaged mitochondria, with suggesting that replacing damaged mitochondria with healthy mitochondria may protect cells from further damage. Moreover, mitochondria can also be actively released into the extracellular space and potentially be transferred between the cells in the central nervous system. This increased interest in mitochondrial therapy calls for a deeper understanding of the mechanisms, which build the basis for mitochondrial transfer, uptake, and cellular defense. In this review, questions related to the involvement of mitochondria in the pathogenesis of cancer will be discussed. Particular attention will be paid to mitochondrial transplantation as a therapeutic approach to treat the mitochondrial dysfunction under some pathological conditions.
Collapse
|
24
|
Hong Q, Chen H, Sun J, Wang C. Memristive Circuit Implementation of a Self-Repairing Network Based on Biological Astrocytes in Robot Application. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2022; 33:2106-2120. [PMID: 33382661 DOI: 10.1109/tnnls.2020.3041624] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A large number of studies have shown that astrocytes can be combined with the presynaptic terminals and postsynaptic spines of neurons to constitute a triple synapse via an endocannabinoid retrograde messenger to achieve a self-repair ability in the human brain. Inspired by the biological self-repair mechanism of astrocytes, this work proposes a self-repairing neuron network circuit that utilizes a memristor to simulate changes in neurotransmitters when a set threshold is reached. The proposed circuit simulates an astrocyte-neuron network and comprises the following: 1) a single-astrocyte-neuron circuit module; 2) an astrocyte-neuron network circuit; 3) a module to detect malfunctions; and 4) a neuron PR (release probability of synaptic transmission) enhancement module. When faults occur in a synapse, the neuron module becomes silent or near silent because of the low PR of the synapses. The circuit can detect faults automatically. The damaged neuron can be repaired by enhancing the PR of other healthy neurons, analogous to the biological repair mechanism of astrocytes. This mechanism helps to repair the damaged circuit. A simulation of the circuit revealed the following: 1) as the number of neurons in the circuit increases, the self-repair ability strengthens and 2) as the number of damaged neurons in the astrocyte-neuron network increases, the self-repair ability weakens, and there is a significant degradation in the performance of the circuit. The self-repairing circuit was used for a robot, and it effectively improved the robots' performance and reliability.
Collapse
|
25
|
Xie HM, Su X, Zhang FY, Dai CL, Wu RH, Li Y, Han XX, Feng XM, Yu B, Zhu SX, Zhou SL. Profile of the RNA in exosomes from astrocytes and microglia using deep sequencing: implications for neurodegeneration mechanisms. Neural Regen Res 2022; 17:608-617. [PMID: 34380901 PMCID: PMC8504369 DOI: 10.4103/1673-5374.320999] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Glial cells play an important role in signal transduction, energy metabolism, extracellular ion homeostasis and neuroprotection of the central nervous system. However, few studies have explained the potential effects of exosomes from glial cells on central nervous system health and disease. In this study, the genes expressed in exosomes from astrocytes and microglia were identified by deep RNA sequencing. Kyoto Encyclopedia of Genes and Genomes analysis indicated that several pathways in these exosomes are responsible for promoting neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease and Huntington’s disease. Gene ontology analysis showed that extracellular exosome, mitochondrion and growth factor activity were enriched in exosomes from the unique astrocyte group, while extracellular exosome and mitochondrion were enriched in exosomes from the unique microglia group. Next, combined with the screening of hub genes, the protein-protein interaction network analysis showed that exosomes from astrocytes influence neurodegenerative diseases through metabolic balance and ubiquitin-dependent protein balance, whereas exosomes from microglia influence neurodegenerative diseases through immune inflammation and oxidative stress. Although there were differences in RNA expression between exosomes from astrocytes and microglia, the groups were related by the hub genes, ubiquitin B and heat shock protein family A (Hsp70) member 8. Ubiquitin B appeared to be involved in pleiotropic regulatory functions, including immune regulation, inflammation inhibition, protein catabolism, intracellular protein transport, exosomes and oxidative stress. The results revealed the clinical significance of exosomes from glia in neurodegenerative diseases. This study was approved by the Animal Ethics Committee of Nantong University, China (approval No. S20180102-152) on January 2, 2018.
Collapse
Affiliation(s)
- Hui-Min Xie
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University; Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xing Su
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Feng-Yuan Zhang
- Medical College of Nantong University, Nantong, Jiangsu Province, China
| | - Chao-Lun Dai
- Medical College of Nantong University, Nantong, Jiangsu Province, China
| | - Rong-Hua Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Xiao Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xing-Mei Feng
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Shun-Xing Zhu
- Laboratory Animals Center, Nantong University, Nantong, Jiangsu Province, China
| | - Song-Lin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
26
|
Nguyen H, Zerimech S, Baltan S. Astrocyte Mitochondria in White-Matter Injury. Neurochem Res 2021; 46:2696-2714. [PMID: 33527218 PMCID: PMC8935665 DOI: 10.1007/s11064-021-03239-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/11/2022]
Abstract
This review summarizes the diverse structure and function of astrocytes to describe the bioenergetic versatility required of astrocytes that are situated at different locations. The intercellular domain of astrocyte mitochondria defines their roles in supporting and regulating astrocyte-neuron coupling and survival against ischemia. The heterogeneity of astrocyte mitochondria, and how subpopulations of astrocyte mitochondria adapt to interact with other glia and regulate axon function, require further investigation. It has become clear that mitochondrial permeability transition pores play a key role in a wide variety of human diseases, whose common pathology may be based on mitochondrial dysfunction triggered by Ca2+ and potentiated by oxidative stress. Reactive oxygen species cause axonal degeneration and a reduction in axonal transport, leading to axonal dystrophies and neurodegeneration including Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease, and Huntington's disease. Developing new tools to allow better investigation of mitochondrial structure and function in astrocytes, and techniques to specifically target astrocyte mitochondria, can help to unravel the role of mitochondrial health and dysfunction in a more inclusive context outside of neuronal cells. Overall, this review will assess the value of astrocyte mitochondria as a therapeutic target to mitigate acute and chronic injury in the CNS.
Collapse
Affiliation(s)
- Hung Nguyen
- Anesthesiology and Peri-Operative Medicine (APOM), Oregon Health and Science University, Portland, OR, 97239, USA
| | - Sarah Zerimech
- Anesthesiology and Peri-Operative Medicine (APOM), Oregon Health and Science University, Portland, OR, 97239, USA
| | - Selva Baltan
- Anesthesiology and Peri-Operative Medicine (APOM), Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
27
|
Claeys W, Van Hoecke L, Lefere S, Geerts A, Verhelst X, Van Vlierberghe H, Degroote H, Devisscher L, Vandenbroucke RE, Van Steenkiste C. The neurogliovascular unit in hepatic encephalopathy. JHEP Rep 2021; 3:100352. [PMID: 34611619 PMCID: PMC8476774 DOI: 10.1016/j.jhepr.2021.100352] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/14/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatic encephalopathy (HE) is a neurological complication of hepatic dysfunction and portosystemic shunting. It is highly prevalent in patients with cirrhosis and is associated with poor outcomes. New insights into the role of peripheral origins in HE have led to the development of innovative treatment strategies like faecal microbiota transplantation. However, this broadening of view has not been applied fully to perturbations in the central nervous system. The old paradigm that HE is the clinical manifestation of ammonia-induced astrocyte dysfunction and its secondary neuronal consequences requires updating. In this review, we will use the holistic concept of the neurogliovascular unit to describe central nervous system disturbances in HE, an approach that has proven instrumental in other neurological disorders. We will describe HE as a global dysfunction of the neurogliovascular unit, where blood flow and nutrient supply to the brain, as well as the function of the blood-brain barrier, are impaired. This leads to an accumulation of neurotoxic substances, chief among them ammonia and inflammatory mediators, causing dysfunction of astrocytes and microglia. Finally, glymphatic dysfunction impairs the clearance of these neurotoxins, further aggravating their effect on the brain. Taking a broader view of central nervous system alterations in liver disease could serve as the basis for further research into the specific brain pathophysiology of HE, as well as the development of therapeutic strategies specifically aimed at counteracting the often irreversible central nervous system damage seen in these patients.
Collapse
Key Words
- ABC, ATP-binding cassette
- ACLF, acute-on-chronic liver failure
- AD, acute decompensation
- ALF, acute liver failure
- AOM, azoxymethane
- AQP4, aquaporin 4
- Acute Liver Failure
- Ammonia
- BBB, blood-brain barrier
- BCRP, breast cancer resistance protein
- BDL, bile duct ligation
- Blood-brain barrier
- Brain edema
- CCL, chemokine ligand
- CCR, C-C chemokine receptor
- CE, cerebral oedema
- CLD, chronic liver disease
- CLDN, claudin
- CNS, central nervous system
- CSF, cerebrospinal fluid
- Cirrhosis
- Energy metabolism
- GS, glutamine synthetase
- Glymphatic system
- HE, hepatic encephalopathy
- HO-1, heme oxygenase 1
- IL-, interleukin
- MMP-9, matrix metalloproteinase 9
- MRP, multidrug resistance associated protein
- NGVU
- NGVU, neurogliovascular unit
- NKCC1, Na-K-2Cl cotransporter 1
- Neuroinflammation
- OCLN, occludin
- ONS, oxidative and nitrosative stress
- Oxidative stress
- P-gp, P-glycoprotein
- PCA, portacaval anastomosis
- PSS, portosystemic shunt
- S1PR2, sphingosine-1-phosphate receptor 2
- SUR1, sulfonylurea receptor 1
- Systemic inflammation
- TAA, thioacetamide
- TGFβ, transforming growth factor beta
- TJ, tight junction
- TNF, tumour necrosis factor
- TNFR1, tumour necrosis factor receptor 1
- ZO, zonula occludens
- mPT, mitochondrial pore transition
Collapse
Affiliation(s)
- Wouter Claeys
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
- Barriers in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- Barriers in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sander Lefere
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences; Liver Research Center Ghent; Ghent University, Ghent, Belgium
| | - Anja Geerts
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
| | - Xavier Verhelst
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
| | - Hans Van Vlierberghe
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
| | - Helena Degroote
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
| | - Lindsey Devisscher
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences; Liver Research Center Ghent; Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- Barriers in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christophe Van Steenkiste
- Antwerp University, Department of Gastroenterology and Hepatology, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| |
Collapse
|
28
|
Singh-Mallah G, Ardalan M, Kang D, Singh K, McMahon CD, Mallard C, Guan J. Administration of cyclic glycine-proline during infancy improves adult spatial memory, astrocyte plasticity, vascularization and GluR-1 expression in rats. Nutr Neurosci 2021; 25:2517-2527. [PMID: 34565308 DOI: 10.1080/1028415x.2021.1980845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cyclic glycine-proline (cGP) is a natural nutrient of breast milk and plays a role in regulating the function of insulin-like growth factor-1 (IGF-1). IGF-1 function is essential for post-natal brain development and adult cognitive function. We evaluated the effects of cGP on spatial memory and histological changes in the hippocampus of the adult rats following infancy administration. Infant rats were treated with either cGP or saline between post-natal days 8 and 22 via oral administration to lactating dams. The spatial memory was evaluated between post-natal days 70 and 75 using Morris water maze tests. The changes of capillaries, astrocytes, synaptophysin and glutamate receptor-1 were examined in the CA1 stratum radiatum of the hippocampus. Compared to saline-treated group, cGP-treated group showed higher path efficiency of entry and lower average heading errors to the platform zone. cGP-treated group also showed longer, larger and more astrocytic processes, more capillaries and higher glutamate receptor-1 expression. The rats made less average heading error to the platform zone have more capillaries, larger and longer astrocytic branches. Thus cGP treatment/supplementation during infancy moderately improved adulthood spatial memory. This long-lasting effect of cGP on memory could be mediated via promoting astrocytic plasticity, vascularization and glutamate trafficking. Therefore, cGP may have a role in regulating IGF-1 function during brain development.
Collapse
Affiliation(s)
- Gagandeep Singh-Mallah
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,AgResearch Ltd., Ruakura Research Centre, Hamilton, New Zealand.,Gravida, National Centre for Growth and Development, Liggins Institute, University of Auckland, Auckland, New Zealand.,Liggins Institute, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Dali Kang
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kuljeet Singh
- AgResearch Ltd., Ruakura Research Centre, Hamilton, New Zealand.,Gravida, National Centre for Growth and Development, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Christopher D McMahon
- AgResearch Ltd., Ruakura Research Centre, Hamilton, New Zealand.,Gravida, National Centre for Growth and Development, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jian Guan
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Gravida, National Centre for Growth and Development, Liggins Institute, University of Auckland, Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
29
|
Yuan J, Fei Y. Lidocaine activates autophagy of astrocytes and ameliorates chronic constriction injury-induced neuropathic pain. J Biochem 2021; 170:25-31. [PMID: 33245112 DOI: 10.1093/jb/mvaa136] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/04/2020] [Indexed: 11/13/2022] Open
Abstract
Lidocaine is a commonly used drug to alleviate neuropathic pain (NP). This work aims to investigate the mechanism of lidocaine in alleviating NP. Chronic constriction injury (CCI) rats were established by surgery to induce NP. We observed the mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) of rats. Immunofluorescence staining was performed to determine the LC3/glial fibrillary acidic protein (GFAP)-positive cells. Rat astrocytes were treated with lipopolysaccharide (LPS) to induce CCI, and then treated with lidocaine or 3-MA (autophagy inhibitor). CCK-8 was performed to detect cell proliferation. Western blot and enzyme-linked immunosorbent assay were performed to detect the level of protein and inflammatory factor. CCI rats exhibited a decrease of MWT and TWL, which was effectively abolished by lidocaine. Lidocaine enhanced the number of LC3/GFAP-positive cells in CCI rats. Moreover, lidocaine inhibited the expression of GFAP and p62, and enhanced LC3-II/LC3-I expression in the LPS-treated astrocytes. Lidocaine inhibited the level of TNF-α and IL-1β in the LPS-treated astrocytes. The influence conferred by lidocaine was effectively abolished by 3-MA. In conclusion, our work demonstrates that lidocaine activates autophagy of astrocytes and ameliorates CCI-induced NP. Thus, our study provides a further experimental basis for the mechanism of lidocaine to alleviate NP.
Collapse
Affiliation(s)
- Jiaqi Yuan
- Department of Anesthesiology, Sir Run Run Shaw Hospital, No. 3 East Qingchun Road, Jianggan District, Hangzhou, 310020 Zhejiang, China
| | - Yue Fei
- Department of Anesthesiology, Sir Run Run Shaw Hospital, No. 3 East Qingchun Road, Jianggan District, Hangzhou, 310020 Zhejiang, China
| |
Collapse
|
30
|
Wen ZH, Huang SY, Kuo HM, Chen CT, Chen NF, Chen WF, Tsui KH, Liu HT, Sung CS. Fumagillin Attenuates Spinal Angiogenesis, Neuroinflammation, and Pain in Neuropathic Rats after Chronic Constriction Injury. Biomedicines 2021; 9:biomedicines9091187. [PMID: 34572376 PMCID: PMC8470034 DOI: 10.3390/biomedicines9091187] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022] Open
Abstract
Introduction: Angiogenesis in the central nervous system is visible in animal models of neuroinflammation and bone cancer pain. However, whether spinal angiogenesis exists and contributes to central sensitization in neuropathic pain remains unclear. This study analyzes the impact of angiogenesis on spinal neuroinflammation in neuropathic pain. Methods: Rats with chronic constriction injury (CCI) to the sciatic nerve underwent the implantation of an intrathecal catheter. Fumagillin or vascular endothelial growth factor-A antibody (anti-VEGF-A) was administered intrathecally. Nociceptive behaviors, cytokine immunoassay, Western blot, and immunohistochemical analysis assessed the effect of angiogenesis inhibition on CCI-induced neuropathic pain. Results: VEGF, cluster of differentiation 31 (CD31), and von Willebrand factor (vWF) expressions increased after CCI in the ipsilateral lumbar spinal cord compared to that in the contralateral side of CCI and control rats from post-operative day (POD) 7 to 28, with a peak at POD 14. Tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 concentrations, but not IL-10 levels, also increased in the ipsilateral spinal cord after CCI. Fumagillin and anti-VEGF-A reduced CCI-induced thermal hyperalgesia from POD 5 to 14 and mechanical allodynia from POD 3 to 14. Fumagillin reduced CCI-upregulated expressions of angiogenic factors and astrocytes. Furthermore, fumagillin decreased TNF-α and IL-6 amounts and increased IL-10 levels at POD 7 and 14, but not IL-1β concentrations. Conclusions: Fumagillin significantly ameliorates CCI-induced nociceptive sensitization, spinal angiogenesis, and astrocyte activation. Our results suggest that angiogenesis inhibitor treatment suppresses peripheral neuropathy-induced central angiogenesis, neuroinflammation, astrocyte activation, and neuropathic pain.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804201, Taiwan; (Z.-H.W.); (H.-M.K.); (C.-T.C.); (W.-F.C.)
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Shi-Ying Huang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China;
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804201, Taiwan; (Z.-H.W.); (H.-M.K.); (C.-T.C.); (W.-F.C.)
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Chao-Ting Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804201, Taiwan; (Z.-H.W.); (H.-M.K.); (C.-T.C.); (W.-F.C.)
| | - Nan-Fu Chen
- Department of Surgery, Division of Neurosurgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 802301, Taiwan;
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804201, Taiwan; (Z.-H.W.); (H.-M.K.); (C.-T.C.); (W.-F.C.)
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
| | - Chun-Sung Sung
- Department of Anesthesiology, Division of Pain Management, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Correspondence: or ; Tel.: +886-2-2875-7549; Fax: +886-2-2875-1597
| |
Collapse
|
31
|
Bisht K, Okojie KA, Sharma K, Lentferink DH, Sun YY, Chen HR, Uweru JO, Amancherla S, Calcuttawala Z, Campos-Salazar AB, Corliss B, Jabbour L, Benderoth J, Friestad B, Mills WA, Isakson BE, Tremblay MÈ, Kuan CY, Eyo UB. Capillary-associated microglia regulate vascular structure and function through PANX1-P2RY12 coupling in mice. Nat Commun 2021; 12:5289. [PMID: 34489419 PMCID: PMC8421455 DOI: 10.1038/s41467-021-25590-8] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 08/17/2021] [Indexed: 12/15/2022] Open
Abstract
Microglia are brain-resident immune cells with a repertoire of functions in the brain. However, the extent of their interactions with the vasculature and potential regulation of vascular physiology has been insufficiently explored. Here, we document interactions between ramified CX3CR1 + myeloid cell somata and brain capillaries. We confirm that these cells are bona fide microglia by molecular, morphological and ultrastructural approaches. Then, we give a detailed spatio-temporal characterization of these capillary-associated microglia (CAMs) comparing them with parenchymal microglia (PCMs) in their morphological activities including during microglial depletion and repopulation. Molecularly, we identify P2RY12 receptors as a regulator of CAM interactions under the control of released purines from pannexin 1 (PANX1) channels. Furthermore, microglial elimination triggered capillary dilation, blood flow increase, and impaired vasodilation that were recapitulated in P2RY12-/- and PANX1-/- mice suggesting purines released through PANX1 channels play important roles in activating microglial P2RY12 receptors to regulate neurovascular structure and function.
Collapse
Affiliation(s)
- Kanchan Bisht
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Kenneth A Okojie
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Kaushik Sharma
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Dennis H Lentferink
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Yu-Yo Sun
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Hong-Ru Chen
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Joseph O Uweru
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Saipranusha Amancherla
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Zainab Calcuttawala
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Antony Brayan Campos-Salazar
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Bruce Corliss
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lara Jabbour
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jordan Benderoth
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Bria Friestad
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - William A Mills
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Département de médecine moléculaire, Université Laval, Québec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Colombia, Vancouver, BC, Canada
| | - Chia-Yi Kuan
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B Eyo
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA.
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
32
|
Telegin GB, Minakov AN, Chernov AS, Kazakov VA, Kalabina EA, Manskikh VN, Asyutin DS, Belogurov AA, Gabibov AG, Konovalov NA, Spallone A. A New Precision Minimally Invasive Method of Glial Scar Simulation in the Rat Spinal Cord Using Cryoapplication. Front Surg 2021; 8:607551. [PMID: 34336912 PMCID: PMC8320592 DOI: 10.3389/fsurg.2021.607551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
According to the World Health Organization, every year worldwide up to 500,000 people suffer a spinal cord injury (SCI). Various animal biomodels are essential for searching for novel protocols and therapeutic approaches for SCI treatment. We have developed an original model of post-traumatic spinal cord glial scarring in rats through cryoapplication. With this method the low-temperature liquid nitrogen is used for the cryodestruction of the spinal cord tissue. Forty-five Sprague Dawley (SD) non-linear male rats of the Specific-pathogen-free (SPF) category were included in this experimental study. A Th13 unilateral hemilaminectomy was performed with dental burr using an operating microscope. A specifically designed cryogenic probe was applied to the spinal cord for one minute through the created bone defect. The animals were euthanized at different time points ranging from 1 to 60 days after cold-induced injury. Their Th12-L1 vertebrae with the injured spinal cord region were removed "en bloc" for histological examination. Our data demonstrate that cryoapplication producing a topical cooling around-20°C, caused a highly standardized transmural lesion of the spinal cord in the dorsoventral direction. The lesion had an "hour-glass" shape on histological sections. During the entire study period (days 1-60 of the post-trauma period), the necrotic processes and the development of the glial scar (lesion evolution) were contained in the surgically approached vertebral space (Th13). Unlike other known experimental methods of SCI simulation (compression, contusion, etc.), the proposed technique is characterized by minimal invasiveness, high precision, and reproducibility. Also, histological findings, lesion size, and postoperative clinical course varied only slightly between different animals. An original design of the cryoprobe used in the study played a primary role in the achieving of these results. The spinal cord lesion's detailed functional morphology is described at different time points (1-60 days) after the produced cryoinjury. Also, changes in the number of macrophages at distinct time points, neoangiogenesis and the formation of the glial scar's fibrous component, including morphodynamic characteristics of its evolution, are analyzed. The proposed method of cryoapplication for inducing reproducible glial scars could facilitate a better understanding of the self-recovery processes in the damaged spinal cord. It would be evidently helpful for finding innovative approaches to the SCI treatment.
Collapse
Affiliation(s)
- Georgii B. Telegin
- Branch of Shemyakin and Ovchinnikov, Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexey N. Minakov
- Branch of Shemyakin and Ovchinnikov, Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Aleksandr S. Chernov
- Branch of Shemyakin and Ovchinnikov, Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vitaly A. Kazakov
- Branch of Shemyakin and Ovchinnikov, Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Elena A. Kalabina
- Branch of Shemyakin and Ovchinnikov, Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vasily N. Manskikh
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry S. Asyutin
- Department of Spinal Neurosurgery, N.N. Burdenko National Scientific and Practical Center for Neurosurgery, RF Health Ministry, Moscow, Russia
| | - Alexey A. Belogurov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexander G. Gabibov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay A. Konovalov
- Department of Spinal Neurosurgery, N.N. Burdenko National Scientific and Practical Center for Neurosurgery, RF Health Ministry, Moscow, Russia
| | - Aldo Spallone
- Department of Clinical Neurosciences, NCL-Neuromed Institute of Neurosciences, Rome, Italy
- Department of Nervous Diseases, RUDN University, Moscow, Russia
| |
Collapse
|
33
|
Burtscher J, Millet GP, Place N, Kayser B, Zanou N. The Muscle-Brain Axis and Neurodegenerative Diseases: The Key Role of Mitochondria in Exercise-Induced Neuroprotection. Int J Mol Sci 2021; 22:6479. [PMID: 34204228 PMCID: PMC8235687 DOI: 10.3390/ijms22126479] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Regular exercise is associated with pronounced health benefits. The molecular processes involved in physiological adaptations to exercise are best understood in skeletal muscle. Enhanced mitochondrial functions in muscle are central to exercise-induced adaptations. However, regular exercise also benefits the brain and is a major protective factor against neurodegenerative diseases, such as the most common age-related form of dementia, Alzheimer's disease, or the most common neurodegenerative motor disorder, Parkinson's disease. While there is evidence that exercise induces signalling from skeletal muscle to the brain, the mechanistic understanding of the crosstalk along the muscle-brain axis is incompletely understood. Mitochondria in both organs, however, seem to be central players. Here, we provide an overview on the central role of mitochondria in exercise-induced communication routes from muscle to the brain. These routes include circulating factors, such as myokines, the release of which often depends on mitochondria, and possibly direct mitochondrial transfer. On this basis, we examine the reported effects of different modes of exercise on mitochondrial features and highlight their expected benefits with regard to neurodegeneration prevention or mitigation. In addition, knowledge gaps in our current understanding related to the muscle-brain axis in neurodegenerative diseases are outlined.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Grégoire P. Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Bengt Kayser
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| |
Collapse
|
34
|
In Vitro Model for Ischemic Stroke: Functional Analysis of Vascular Smooth Muscle Cells. Cell Mol Neurobiol 2021; 42:2289-2304. [PMID: 34032948 DOI: 10.1007/s10571-021-01103-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
The Neurovascular Unit (NVU) is formed by vascular and neural cells controlling the cerebral hyperaemia. All the components are anatomically and functionally linked to each other, resulting in a highly efficient regulation of the cerebral blood flow, which, when interrupted, can lead to stroke. An ischemic stroke (IS) is the most common type of stroke with high rates of morbidity, mortality and disability. Therefore, it is of extreme importance to protect the functional and structural integrity of the NVU in patients with IS, understanding the mechanisms involved and how it affects each component of the NVU. Thus, the aim of this work is to analyse how the vascular smooth muscle cells from the rat middle cerebral artery function/react after an ischemic event. To mimic this event, primary cortical cultures were challenged to oxygen and glucose deprivation (OGD) for 4 h and 6 h, and the smooth muscle cells (SMCs) contractility was analysed after exposure to different media previously conditioned by the cortical cultures upon reperfusion. The results show a dual effect on the SMCs response to the vasorelaxant agent, only for cells exposed to the reperfusion media conditioned by neuron-glia cultures challenged by OGD, leading to increased relaxation of the SMCs for OGD 4 h, whereas for OGD 6 h the effect is reversed leading to contraction of the SMCs. These differences demonstrate that the astrocytes mediate the vasoactive response of vascular smooth muscle by releasing factors into the reperfusion medium, and the hypoxia time is fundamental for a beneficial/harmful response by the vascular smooth muscle.
Collapse
|
35
|
Oyefeso FA, Muotri AR, Wilson CG, Pecaut MJ. Brain organoids: A promising model to assess oxidative stress-induced central nervous system damage. Dev Neurobiol 2021; 81:653-670. [PMID: 33942547 DOI: 10.1002/dneu.22828] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
Oxidative stress (OS) is one of the most significant propagators of systemic damage with implications for widespread pathologies such as vascular disease, accelerated aging, degenerative disease, inflammation, and traumatic injury. OS can be induced by numerous factors such as environmental conditions, lifestyle choices, disease states, and genetic susceptibility. It is tied to the accumulation of free radicals, mitochondrial dysfunction, and insufficient antioxidant protection, which leads to cell aging and tissue degeneration over time. Unregulated systemic increase in reactive species, which contain harmful free radicals, can lead to diverse tissue-specific OS responses and disease. Studies of OS in the brain, for example, have demonstrated how this state contributes to neurodegeneration and altered neural plasticity. As the worldwide life expectancy has increased over the last few decades, the prevalence of OS-related diseases resulting from age-associated progressive tissue degeneration. Unfortunately, vital translational research studies designed to identify and target disease biomarkers in human patients have been impeded by many factors (e.g., limited access to human brain tissue for research purposes and poor translation of experimental models). In recent years, stem cell-derived three-dimensional tissue cultures known as "brain organoids" have taken the spotlight as a novel model for studying central nervous system (CNS) diseases. In this review, we discuss the potential of brain organoids to model the responses of human neural cells to OS, noting current and prospective limitations. Overall, brain organoids show promise as an innovative translational model to study CNS susceptibility to OS and elucidate the pathophysiology of the aging brain.
Collapse
Affiliation(s)
- Foluwasomi A Oyefeso
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Alysson R Muotri
- Department of Pediatrics/Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Christopher G Wilson
- Lawrence D. Longo, MD, Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Michael J Pecaut
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
36
|
Hayashida K, Takegawa R, Shoaib M, Aoki T, Choudhary RC, Kuschner CE, Nishikimi M, Miyara SJ, Rolston DM, Guevara S, Kim J, Shinozaki K, Molmenti EP, Becker LB. Mitochondrial transplantation therapy for ischemia reperfusion injury: a systematic review of animal and human studies. J Transl Med 2021; 19:214. [PMID: 34001191 PMCID: PMC8130169 DOI: 10.1186/s12967-021-02878-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/07/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Mitochondria are essential organelles that provide energy for cellular functions, participate in cellular signaling and growth, and facilitate cell death. Based on their multifactorial roles, mitochondria are also critical in the progression of critical illnesses. Transplantation of mitochondria has been reported as a potential promising approach to treat critical illnesses, particularly ischemia reperfusion injury (IRI). However, a systematic review of the relevant literature has not been conducted to date. Here, we systematically reviewed the animal and human studies relevant to IRI to summarize the evidence for mitochondrial transplantation. METHODS We searched MEDLINE, the Cochrane library, and Embase and performed a systematic review of mitochondrial transplantation for IRI in both preclinical and clinical studies. We developed a search strategy using a combination of keywords and Medical Subject Heading/Emtree terms. Studies including cell-mediated transfer of mitochondria as a transfer method were excluded. Data were extracted to a tailored template, and data synthesis was descriptive because the data were not suitable for meta-analysis. RESULTS Overall, we identified 20 animal studies and two human studies. Among animal studies, 14 (70%) studies focused on either brain or heart IRI. Both autograft and allograft mitochondrial transplantation were used in 17 (85%) animal studies. The designs of the animal studies were heterogeneous in terms of the route of administration, timing of transplantation, and dosage used. Twelve (60%) studies were performed in a blinded manner. All animal studies reported that mitochondrial transplantation markedly mitigated IRI in the target tissues, but there was variation in biological biomarkers and pathological changes. The human studies were conducted with a single-arm, unblinded design, in which autologous mitochondrial transplantation was applied to pediatric patients who required extracorporeal membrane oxygenation (ECMO) for IRI-associated myocardial dysfunction after cardiac surgery. CONCLUSION The evidence gathered from our systematic review supports the potential beneficial effects of mitochondrial transplantation after IRI, but its clinical translation remains limited. Further investigations are thus required to explore the mechanisms of action and patient outcomes in critical settings after mitochondrial transplantation. Systematic review registration The study was registered at UMIN under the registration number UMIN000043347.
Collapse
Affiliation(s)
- Kei Hayashida
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA. .,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA.
| | - Ryosuke Takegawa
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Muhammad Shoaib
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA.,Zucker School of Medicine At Hofstra/Northwell, New York, NY, USA
| | - Tomoaki Aoki
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Rishabh C Choudhary
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Cyrus E Kuschner
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA.,Zucker School of Medicine At Hofstra/Northwell, New York, NY, USA
| | - Mitsuaki Nishikimi
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Santiago J Miyara
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Daniel M Rolston
- Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA.,Zucker School of Medicine At Hofstra/Northwell, New York, NY, USA
| | - Sara Guevara
- Department of Surgery, Northwell Health, Manhasset, NY, USA
| | - Junhwan Kim
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA.,Zucker School of Medicine At Hofstra/Northwell, New York, NY, USA
| | - Koichiro Shinozaki
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA.,Zucker School of Medicine At Hofstra/Northwell, New York, NY, USA
| | - Ernesto P Molmenti
- Zucker School of Medicine At Hofstra/Northwell, New York, NY, USA.,Department of Surgery, Northwell Health, Manhasset, NY, USA
| | - Lance B Becker
- The Feinstein Institutes for Medical Research, Northwell Health System, 350 Community Drive, Manhasset, NY, USA.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, 350 Community Dr, Manhasset, NY, 11030, USA.,Zucker School of Medicine At Hofstra/Northwell, New York, NY, USA
| |
Collapse
|
37
|
Cameron T, Bennet T, Rowe EM, Anwer M, Wellington CL, Cheung KC. Review of Design Considerations for Brain-on-a-Chip Models. MICROMACHINES 2021; 12:441. [PMID: 33921018 PMCID: PMC8071412 DOI: 10.3390/mi12040441] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
In recent years, the need for sophisticated human in vitro models for integrative biology has motivated the development of organ-on-a-chip platforms. Organ-on-a-chip devices are engineered to mimic the mechanical, biochemical and physiological properties of human organs; however, there are many important considerations when selecting or designing an appropriate device for investigating a specific scientific question. Building microfluidic Brain-on-a-Chip (BoC) models from the ground-up will allow for research questions to be answered more thoroughly in the brain research field, but the design of these devices requires several choices to be made throughout the design development phase. These considerations include the cell types, extracellular matrix (ECM) material(s), and perfusion/flow considerations. Choices made early in the design cycle will dictate the limitations of the device and influence the end-point results such as the permeability of the endothelial cell monolayer, and the expression of cell type-specific markers. To better understand why the engineering aspects of a microfluidic BoC need to be influenced by the desired biological environment, recent progress in microfluidic BoC technology is compared. This review focuses on perfusable blood-brain barrier (BBB) and neurovascular unit (NVU) models with discussions about the chip architecture, the ECM used, and how they relate to the in vivo human brain. With increased knowledge on how to make informed choices when selecting or designing BoC models, the scientific community will benefit from shorter development phases and platforms curated for their application.
Collapse
Affiliation(s)
- Tiffany Cameron
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Tanya Bennet
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Elyn M. Rowe
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Mehwish Anwer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Karen C. Cheung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Electrical & Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
38
|
Zhao Y, Wang S, Song X, Yuan J, Qi D, Gu X, Yin MY, Han Z, Zhu Y, Liu Z, Zhang Y, Wei L, Wei ZZ. Glial Cell-Based Vascular Mechanisms and Transplantation Therapies in Brain Vessel and Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:627682. [PMID: 33841101 PMCID: PMC8032950 DOI: 10.3389/fncel.2021.627682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodevelopmental and neurodegenerative diseases (NDDs) with severe neurological/psychiatric symptoms, such as cerebrovascular pathology in AD, CAA, and chronic stroke, have brought greater attention with their incidence and prevalence having markedly increased over the past few years. Causes of the significant neuropathologies, especially those observed in neurological diseases in the CNS, are commonly believed to involve multiple factors such as an age, a total environment, genetics, and an immunity contributing to their progression, neuronal, and vascular injuries. We primarily focused on the studies of glial involvement/dysfunction in part with the blood-brain barrier (BBB) and the neurovascular unit (NVU) changes, and the vascular mechanisms, which have been both suggested as critical roles in chronic stroke and many other NDDs. It has been noted that glial cells including astrocytes (which outnumber other cell types in the CNS) essentially contribute more to the BBB integrity, extracellular homeostasis, neurotransmitter release, regulation of neurogenic niches in response to neuroinflammatory stimulus, and synaptic plasticity. In a recent study for NDDs utilizing cellular and molecular biology and genetic and pharmacological tools, the role of reactive astrocytes (RACs) and gliosis was demonstrated, able to trigger pathophysiological/psychopathological detrimental changes during the disease progression. We speculate, in particular, the BBB, the NVU, and changes of the astrocytes (potentially different populations from the RACs) not only interfere with neuronal development and synaptogenesis, but also generate oxidative damages, contribute to beta-amyloid clearances and disrupted vasculature, as well as lead to neuroinflammatory disorders. During the past several decades, stem cell therapy has been investigated with a research focus to target related neuro-/vascular pathologies (cell replacement and repair) and neurological/psychiatric symptoms (paracrine protection and homeostasis). Evidence shows that transplantation of neurogenic or vasculogenic cells could be achieved to pursue differentiation and maturation within the diseased brains as expected. It would be hoped that, via regulating functions of astrocytes, astrocytic involvement, and modulation of the BBB, the NVU and astrocytes should be among major targets for therapeutics against NDDs pathogenesis by drug and cell-based therapies. The non-invasive strategies in combination with stem cell transplantation such as the well-tested intranasal deliveries for drug and stem cells by our and many other groups show great translational potentials in NDDs. Neuroimaging and clinically relevant analyzing tools need to be evaluated in various NDDs brains.
Collapse
Affiliation(s)
- Yingying Zhao
- Beijing Clinical Research Institute, Beijing, China.,Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States.,Department of Critical Care Medicine, Airport Hospital of Tianjin Medical University General Hospital, Tianjin, China
| | - Shuanglin Wang
- Department of Critical Care Medicine, Airport Hospital of Tianjin Medical University General Hospital, Tianjin, China.,Department of Cardiovascular Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China.,Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaopeng Song
- Mclean Imaging Center, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| | - Junliang Yuan
- Mclean Imaging Center, Harvard Medical School, McLean Hospital, Belmont, MA, United States.,Department of Neurology, Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
| | - Dong Qi
- Beijing Clinical Research Institute, Beijing, China
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Michael Yaoyao Yin
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States.,Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Zhou Han
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, United States
| | - Yanbing Zhu
- Beijing Clinical Research Institute, Beijing, China
| | - Zhandong Liu
- Beijing Clinical Research Institute, Beijing, China
| | - Yongbo Zhang
- Beijing Clinical Research Institute, Beijing, China
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States.,Emory Specialized Center of Sex Differences, Emory University, Atlanta, GA, United States
| |
Collapse
|
39
|
Park JH, Hayakawa K. Extracellular Mitochondria Signals in CNS Disorders. Front Cell Dev Biol 2021; 9:642853. [PMID: 33748135 PMCID: PMC7973090 DOI: 10.3389/fcell.2021.642853] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/26/2021] [Indexed: 01/01/2023] Open
Abstract
Mitochondria actively participate in the regulation of cell respiratory mechanisms, metabolic processes, and energy homeostasis in the central nervous system (CNS). Because of the requirement of high energy, neuronal functionality and viability are largely dependent on mitochondrial functionality. In the context of CNS disorders, disruptions of metabolic homeostasis caused by mitochondrial dysfunction lead to neuronal cell death and neuroinflammation. Therefore, restoring mitochondrial function becomes a primary therapeutic target. Recently, accumulating evidence suggests that active mitochondria are secreted into the extracellular fluid and potentially act as non-cell-autonomous signals in CNS pathophysiology. In this mini-review, we overview findings that implicate the presence of cell-free extracellular mitochondria and the critical role of intercellular mitochondrial transfer in various rodent models of CNS disorders. We also discuss isolated mitochondrial allograft as a novel therapeutic intervention for CNS disorders.
Collapse
Affiliation(s)
- Ji-Hyun Park
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
40
|
González-Molina LA, Villar-Vesga J, Henao-Restrepo J, Villegas A, Lopera F, Cardona-Gómez GP, Posada-Duque R. Extracellular Vesicles From 3xTg-AD Mouse and Alzheimer's Disease Patient Astrocytes Impair Neuroglial and Vascular Components. Front Aging Neurosci 2021; 13:593927. [PMID: 33679370 PMCID: PMC7933224 DOI: 10.3389/fnagi.2021.593927] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Astrocytes are specialized glial cells that are essential components of the neurovascular unit (NVU) and are involved in neurodevelopment, brain maintenance and repair, and neurodegeneration. Astrocytes mediate these processes by releasing cellular mediators such as extracellular vesicles (EVs). EVs are vehicles of cell-cell communication and have been proposed as mediators of damage in AD. However, the transcellular mechanism by which Alzheimer disease (AD) astrocytes impair the function of NVU components is poorly understood. Therefore, we evaluated the effects of adult PS1-KI and 3xTg-AD astrocyte conditioned media (CM) and EVs on NVU components (neuroglia and endothelium) in vitro. Additionally, SAD and FAD astrocyte-derived EVs (A-EVs) were characterized, and we evaluated their effects on NVU in cocultured cells in vitro and on intrahippocampal CA1 cells in vivo. Surprisingly, cultured 3xTg-AD astrocytes showed increased glial fibrillary acidic protein (GFAP) reactivity compared to PS1-KI astrocytes, which denotes astrocytic hyperreactivity. CM from adult mice 3xTg-AD astrocytes increased cell-cell gaps between endothelial cells, filopodia-like dendritic protrusions in neurons and neuronal and endothelial cell death. 3xTg-AD A-EVs induced neurotoxicity and increased astrocyte GFAP reactivity. Cultured human postmortem astrocytes from AD patients also increased GFAP reactivity and EVs release. No differences in the size or number of A-EVs were detected between AD and control samples; however, both SAD and FAD A-EVs showed increased expression of the surface marker aquaporin 4. A-EVs induced cytotoxicity and astrocyte hyperactivation: specifically, FAD A-EVs induced neuroglial cytotoxicity and increased gaps between the endothelium, while SAD A-EVs mainly altered the endothelium. Similarly, both AD A-EVs increased astrocyte GS reactivity and vascular deterioration in vivo. We associated this finding with perivascular reactive astrocytes and vascular deterioration in the human AD brain. In summary, these results suggest that AD A-EVs impair neuroglial and vascular components.
Collapse
Affiliation(s)
- Luis Alfonso González-Molina
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia.,Facultad de Ciencias Exactas y Naturales, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| | - Juan Villar-Vesga
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia.,Facultad de Ciencias Exactas y Naturales, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| | - Julián Henao-Restrepo
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia.,Facultad de Ciencias Exactas y Naturales, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| | - Andrés Villegas
- Neurobank, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellin, Colombia
| | - Francisco Lopera
- Neurobank, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia, Medellin, Colombia
| | - Gloria Patricia Cardona-Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia
| | - Rafael Posada-Duque
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin, Colombia.,Facultad de Ciencias Exactas y Naturales, Instituto de Biología, Universidad de Antioquia, Medellin, Colombia
| |
Collapse
|
41
|
Wang HK, Huang CY, Chen YW, Sun YT. Hyperglycemia compromises the ischemia-provoked dedifferentiation of cerebral pericytes through p21-SOX2 signaling in high-fat diet-induced murine model. Diab Vasc Dis Res 2021; 18:1479164121990641. [PMID: 33557613 PMCID: PMC8482726 DOI: 10.1177/1479164121990641] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM Diabetes-related cerebral small vessel disease (CSVD) causes neurological deficits. Patients with diabetes showed pericyte loss as a hallmark of retinopathy. Cerebral pericytes, which densely localize around brain capillaries, are quiescent stem cells regulating regeneration of brain and may have a role in CSVD development. This study investigated whether diabetes impairs ischemia-provoked dedifferentiation of pericytes. METHODS A murine high-fat diet (HFD)-induced diabetes model was used. After cerebral ischemia induction in the mice, pericytes were isolated and grown for a sphere formation assay. RESULTS The sphere counts from the HFD group were lower than those in the chow group. As the spheres formed, pericyte marker levels decreased and SOX2 levels increased gradually in the chow group, but not in the HFD group. Before sphere formation, pericytes from the HFD group showed high p21 levels. The use of a p21 inhibitor rescued the reduction of sphere counts in the HFD group. At cellular level, hyperglycemia-induced ROS increased the level of p21 in cerebral pericytes. The p21-SOX2 signaling was then activated after oxygen-glucose deprivation. CONCLUSION HFD-induced diabetes compromises the stemness of cerebral pericytes by altering p21-SOX2 signaling. These results provide evidence supporting the role of pericytes in diabetes-related CSVD and subsequent cerebral dysfunction.
Collapse
Affiliation(s)
- Hao-Kuang Wang
- School of Medicine for International Students, I-Shou University, Kaohsiung
- Department of Neurosurgery, E-Da Hospital, I-Shou University, Kaohsiung
| | - Chih-Yuan Huang
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Yun-Wen Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan
| | - Yuan-Ting Sun
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
- Department of Genomic Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| |
Collapse
|
42
|
Finetti F, Travelli C, Ercoli J, Colombo G, Buoso E, Trabalzini L. Prostaglandin E2 and Cancer: Insight into Tumor Progression and Immunity. BIOLOGY 2020; 9:E434. [PMID: 33271839 PMCID: PMC7760298 DOI: 10.3390/biology9120434] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022]
Abstract
The involvement of inflammation in cancer progression has been the subject of research for many years. Inflammatory milieu and immune response are associated with cancer progression and recurrence. In different types of tumors, growth and metastatic phenotype characterized by the epithelial mesenchymal transition (EMT) process, stemness, and angiogenesis, are increasingly associated with intrinsic or extrinsic inflammation. Among the inflammatory mediators, prostaglandin E2 (PGE2) supports epithelial tumor aggressiveness by several mechanisms, including growth promotion, escape from apoptosis, transactivation of tyrosine kinase growth factor receptors, and induction of angiogenesis. Moreover, PGE2 is an important player in the tumor microenvironment, where it suppresses antitumor immunity and regulates tumor immune evasion, leading to increased tumoral progression. In this review, we describe the current knowledge on the pro-tumoral activity of PGE2 focusing on its role in cancer progression and in the regulation of the tumor microenvironment.
Collapse
Affiliation(s)
- Federica Finetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, 27100 Pavia, Italy; (C.T.); (E.B.)
| | - Jasmine Ercoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| | - Giorgia Colombo
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Erica Buoso
- Department of Pharmaceutical Sciences, University of Pavia, 27100 Pavia, Italy; (C.T.); (E.B.)
| | - Lorenza Trabalzini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| |
Collapse
|
43
|
Cai J, Yan Y, Zhang D, Zhu M, Wang Z, Zhang X, Xu M. Silencing of lncRNA Gm14461 alleviates pain in trigeminal neuralgia through inhibiting astrocyte activation. IUBMB Life 2020; 72:2663-2671. [PMID: 33141516 DOI: 10.1002/iub.2395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/16/2020] [Accepted: 09/29/2020] [Indexed: 02/02/2023]
Abstract
Our previous study showed that silencing of lncRNA Gm14461 alleviated pain in a murine model of trigeminal neuralgia (TN), but the molecular mechanism remains not fully understood. Evidence indicates that astrocyte activation and autophagy are involved in the development of TN. Herein, this study aimed to elucidate whether the pain-relief effect of Gm14461 silencing in TN involved regulation of astrocyte activation and autophagy. A murine model of TN was induced by chronic constriction injury of the infraorbital nerve surgery. The mechanical withdrawal threshold (MWT) was measured to assess the analgesic effect of Gm14461 silencing. Mouse astrocytes were treated with lipopolysaccharide (LPS) as a cell model. Astrocyte activation was evaluated by glial fibrillary acidic protein (GFAP) immunofluorescence and western blot analysis of GFAP. Autophagy was evaluated by LC3 immunofluorescence and western blot analysis of autophagy-related proteins. The results showed that Gm14461 silencing increased MWT value in TN model mice. Meanwhile, Gm14461 silencing inhibited astrocyte activation and enhanced autophagy in both TN mice and LPS-treated astrocytes. The enhancement of autophagy by Gm14461 silencing involved the activation of the AMPK signaling and the suppression of the Akt/mTOR signaling. Collectively, the analgesic effect of Gm14461 silencing in TN was related to attenuation of astrocyte activation via enhancement of autophagy.
Collapse
Affiliation(s)
- Jun Cai
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Yan
- Department of Pain Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Daying Zhang
- Department of Pain Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Mengye Zhu
- Department of Pain Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhijian Wang
- Department of Pain Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xuexue Zhang
- Department of Pain Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Mu Xu
- Department of Pain Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
44
|
Shanmughapriya S, Langford D, Natarajaseenivasan K. Inter and Intracellular mitochondrial trafficking in health and disease. Ageing Res Rev 2020; 62:101128. [PMID: 32712108 DOI: 10.1016/j.arr.2020.101128] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
Neurons and glia maintain central nervous system (CNS) homeostasis through diverse mechanisms of intra- and intercellular signaling. Some of these interactions include the exchange of soluble factors between cells via direct cell-to-cell contact for both short and long-distance transfer of biological materials. Transcellular transfer of mitochondria has emerged as a key example of this communication. This transcellular transfer of mitochondria are dynamically involved in the cellular and tissue response to CNS injury and play beneficial roles in recovery. This review highlights recent research addressing the cause and effect of intra- and intercellular mitochondrial transfer with a specific focus on the future of mitochondrial transplantation therapy. We believe that mitochondrial transfer plays a crucial role during bioenergetic crisis/deficit, but the quality, quantity and mode of mitochondrial transfer determines the protective capacity for the receiving cells. Mitochondrial transplantation is a new treatment paradigm and will overcome the major bottleneck of traditional approach of correcting mitochondria-related disorders.
Collapse
|
45
|
The Role of Neurovascular System in Neurodegenerative Diseases. Mol Neurobiol 2020; 57:4373-4393. [PMID: 32725516 DOI: 10.1007/s12035-020-02023-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022]
Abstract
The neurovascular system (NVS), which consisted of neurons, glia, and vascular cells, is a functional and structural unit of the brain. The NVS regulates blood-brain barrier (BBB) permeability and cerebral blood flow (CBF), thereby maintaining the brain's microenvironment for normal functioning, neuronal survival, and information processing. Recent studies have highlighted the role of vascular dysfunction in several neurodegenerative diseases. This is not unexpected since both nervous and vascular systems are functionally interdependent and show close anatomical apposition, as well as similar molecular pathways. However, despite extensive research, the precise mechanism by which neurovascular dysfunction contributes to neurodegeneration remains incomplete. Therefore, understanding the mechanisms of neurovascular dysfunction in disease conditions may allow us to develop potent and effective therapies for prevention and treatment of neurodegenerative diseases. This review article summarizes the current research in the context of neurovascular signaling associated with neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). We also discuss the potential implication of neurovascular factor as a novel therapeutic target and prognostic marker in patients with neurodegenerative conditions. Graphical Abstract.
Collapse
|
46
|
Taylor X, Cisternas P, You Y, You Y, Xiang S, Marambio Y, Zhang J, Vidal R, Lasagna-Reeves CA. A1 reactive astrocytes and a loss of TREM2 are associated with an early stage of pathology in a mouse model of cerebral amyloid angiopathy. J Neuroinflammation 2020; 17:223. [PMID: 32711525 PMCID: PMC7382050 DOI: 10.1186/s12974-020-01900-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cerebral amyloid angiopathy (CAA) is typified by the cerebrovascular deposition of amyloid. The mechanisms underlying the contribution of CAA to neurodegeneration are not currently understood. Although CAA is highly associated with the accumulation of amyloid beta (Aβ), other amyloids are known to associate with the vasculature. Alzheimer’s disease (AD) is characterized by parenchymal Aβ deposition, intracellular accumulation of tau, and significant neuroinflammation. CAA increases with age and is present in 85–95% of individuals with AD. A substantial amount of research has focused on understanding the connection between parenchymal amyloid and glial activation and neuroinflammation, while associations between vascular amyloid pathology and glial reactivity remain understudied. Methods Here, we dissect the glial and immune responses associated with early-stage CAA with histological, biochemical, and gene expression analyses in a mouse model of familial Danish dementia (FDD), a neurodegenerative disease characterized by the vascular accumulation of Danish amyloid (ADan). Findings observed in this CAA mouse model were complemented with primary culture assays. Results We demonstrate that early-stage CAA is associated with dysregulation in immune response networks and lipid processing, severe astrogliosis with an A1 astrocytic phenotype, and decreased levels of TREM2 with no reactive microgliosis. Our results also indicate how cholesterol accumulation and ApoE are associated with vascular amyloid deposits at the early stages of pathology. We also demonstrate A1 astrocytic mediation of TREM2 and microglia homeostasis. Conclusion The initial glial response associated with early-stage CAA is characterized by the upregulation of A1 astrocytes without significant microglial reactivity. Gene expression analysis revealed that several AD risk factors involved in immune response and lipid processing may also play a preponderant role in CAA. This study contributes to the increasing evidence that brain cholesterol metabolism, ApoE, and TREM2 signaling are major players in the pathogenesis of AD-related dementias, including CAA. Understanding the basis for possible differential effects of glial response, ApoE, and TREM2 signaling on parenchymal plaques versus vascular amyloid deposits provides important insight for developing future therapeutic interventions.
Collapse
Affiliation(s)
- Xavier Taylor
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Pablo Cisternas
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yanwen You
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yingjian You
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shunian Xiang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yamil Marambio
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Ruben Vidal
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cristian A Lasagna-Reeves
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA. .,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
47
|
Structural and chemical changes in glial cells in the rat neocortex induced by constant occlusion of the middle cerebral artery. Acta Histochem 2020; 122:151573. [PMID: 32622419 DOI: 10.1016/j.acthis.2020.151573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/09/2020] [Accepted: 06/04/2020] [Indexed: 12/21/2022]
Abstract
Stroke-induced changes in neuroglia determine the basic conditions for the survival and damage of neurons in the ischemic core. Here, we studied the immunolocalization of glial cell line-derived neurotrophic factor (GDNF), glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule 1 (Iba-1), and S-100β in the rat parietal cortex after constant occlusion of the middle cerebral artery. These cytoplasmic proteins are specific for different glial cell types. They are used as indicators of activated microglia and astrocytes in immunocytochemical studies. The distribution pattern of all markers changed dramatically with time. GFAP- and S-100β-positive astrocytes were observed in the penumbra zone and marked its boundaries. In days 1-8 after surgery, in the ischemic core, the number of S-100β-immunoreactive astrocytes decreased, and individual pyramidal cells appeared. S-100β-expressing pyramidal cells were localized in cortical layers III and V. They were only found in the ischemic core. Their proportion to the total number of cells was 37.3 ± 3.9 %, 22.2 ± 1.2 %, 16.3 ± 2.3 %, and 5.4 ± 0.3 % on days 1, 3, 8, and 14 after surgery. On day 21, no S-100β-expressing pyramidal cells were observed. The spatial density of GFAP- and S-100β-positive astrocytes increased in the penumbra region adjacent to the ischemic core and decreased in the penumbral periphery. As a result, the width of the perifocal penumbra zone decreased substantially at later stages of the stroke. In the penumbra, on days 1-3 after ischemic injury, GDNF immunoreactivity was mainly localized in neurons, while later on (days 8-21) it was mainly constrained to astrocyte glia. In intact rats, GDNF-positive neurons were situated in cortical layers II/III and V/VI and made up 52 ± 4.5 % of the total neuron population. Their proportion to the total number of neurons was 29 ± 2.1 %, 13.8 ± 0.6 %, and 3.1 ± 0.2 % on days 1, 8, and 21 after surgery. The number of GDNF-positive astrocytes, on the opposite, increased with time after ischemic injury. Iba-1-reactive microglia was mainly localized to the ischemic core. Microglial cells appeared activated as evidenced by their increased size and decreased number of processes and branching density. The spatial density of microglia reached a peak on day 8 after ischemic injury both in the ischemic core and penumbra. An increase in the number of Iba-1-reactive microglia in the ischemic core correlated with a decrease of the number of GFAP-positive astrocytes. The results are discussed in the context of participation of neuroglia in regulation of various neuroprotective and destructive processes.
Collapse
|
48
|
Huang L, Applegate Ii RL, Applegate PM, Gong L, Ocak U, Boling W, Zhang JH. Inhalation of high-concentration hydrogen gas attenuates cognitive deficits in a rat model of asphyxia induced-cardiac arrest. Med Gas Res 2020; 9:122-126. [PMID: 31552874 PMCID: PMC6779004 DOI: 10.4103/2045-9912.266986] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cognitive deficits are a devastating neurological outcome seen in survivors of cardiac arrest. We previously reported water electrolysis derived 67% hydrogen gas inhalation has some beneficial effects on short-term outcomes in a rat model of global brain hypoxia-ischemia induced by asphyxia cardiac arrest. In the present study, we further investigated its protective effects in long-term spatial learning memory function using the same animal model. Water electrolysis derived 67% hydrogen gas was either administered 1 hour prior to cardiac arrest for 1 hour and at 1-hour post-resuscitation for 1 hour (pre- & post-treatment) or at 1-hour post-resuscitation for 2 hours (post-treatment). T-maze and Morris water maze were used for hippocampal memory function evaluation at 7 and 14 days post-resuscitation, respectively. Neuronal degeneration within hippocampal Cornu Ammonis 1 (CA1) regions was examined by Fluoro-Jade staining ex vivo. Hippocampal deficits were detected at 7 and 18 days post-resuscitation, with increased neuronal degeneration within hippocampal CA1 regions. Both hydrogen gas treatment regimens significantly improved spatial learning function and attenuated neuronal degeneration within hippocampal CA1 regions at 18 days post-resuscitation. Our findings suggest that water electrolysis derived 67% hydrogen gas may be an effective therapeutic approach for improving cognitive outcomes associated with global brain hypoxia-ischemia following cardiac arrest. The study was approved by the Animal Health and Safety Committees of Loma Linda University, USA (approval number: IACUC #8170006) on March 2, 2017.
Collapse
Affiliation(s)
- Lei Huang
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Richard L Applegate Ii
- Department of Anesthesiology and Pain Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Patricia M Applegate
- Department of Cardiology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Lei Gong
- Department of Pharmacy, 1st Affiliated Hospital to Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Umut Ocak
- Department of Basic Sciences, Division of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Warren Boling
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
49
|
Mason S. A Novel, Multi-Faceted Perception of Lactate in Neurology. Front Neurosci 2020; 14:460. [PMID: 32499676 PMCID: PMC7242720 DOI: 10.3389/fnins.2020.00460] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 04/15/2020] [Indexed: 12/20/2022] Open
Affiliation(s)
- Shayne Mason
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| |
Collapse
|
50
|
Gollihue J, Norris C. Astrocyte mitochondria: Central players and potential therapeutic targets for neurodegenerative diseases and injury. Ageing Res Rev 2020; 59:101039. [PMID: 32105849 DOI: 10.1016/j.arr.2020.101039] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/23/2020] [Accepted: 02/23/2020] [Indexed: 01/16/2023]
Abstract
Mitochondrial function has long been the focus of many therapeutic strategies for ameliorating age-related neurodegeneration and cognitive decline. Historically, the role of mitochondria in non-neuronal cell types has been overshadowed by neuronal mitochondria, which are responsible for the bulk of oxidative metabolism in the brain. Despite this neuronal bias, mitochondrial function in glial cells, particularly astrocytes, is increasingly recognized to play crucial roles in overall brain metabolism, synaptic transmission, and neuronal protection. Changes in astrocytic mitochondrial function appear to be intimately linked to astrocyte activation/reactivity found in most all age-related neurodegenerative diseases. Here, we address the importance of mitochondrial function to astrocyte signaling and consider how mitochondria could contribute to both the detrimental and protective properties of activated astrocytes. Strategies for protecting astrocytic mitochondrial function, promoting bidirectional transfer of mitochondria between astrocytes and neurons, and transplanting healthy mitochondria to diseased nervous tissue are also discussed.
Collapse
|