1
|
Zeng M, Peng M, Liang J, Sun H. The Role of Gut Microbiota in Blood-Brain Barrier Disruption after Stroke. Mol Neurobiol 2024; 61:9735-9755. [PMID: 37498481 DOI: 10.1007/s12035-023-03512-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Growing evidence has proved that alterations in the gut microbiota have been linked to neurological disorders including stroke. Structural and functional disruption of the blood-brain barrier (BBB) is observed after stroke. In this context, there is pioneering evidence supporting that gut microbiota may be involved in the pathogenesis of stroke by regulating the BBB function. However, only a few experimental studies have been performed on stroke models to observe the BBB by altering the structure of gut microbiota, which warrant further exploration. Therefore, in order to provide a novel mechanism for stroke and highlight new insights into BBB modification as a stroke intervention, this review summarizes existing evidence of the relationship between gut microbiota and BBB integrity and discusses the mechanisms of gut microbiota on BBB dysfunction and its role in stroke.
Collapse
Affiliation(s)
- Meiqin Zeng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Meichang Peng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jianhao Liang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Lombardi AM, Wong H, Bower ME, Milstead R, Borski C, Schmitt E, Griffioen M, LaPlante L, Ehringer MA, Stitzel J, Hoeffer CA. AKT2 modulates astrocytic nicotine responses in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596856. [PMID: 38854016 PMCID: PMC11160815 DOI: 10.1101/2024.05.31.596856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
A better understanding of nicotine neurobiology is needed to reduce or prevent chronic addiction, ameliorate the detrimental effects of nicotine withdrawal, and increase successful cessation of use. Nicotine binds and activates two astrocyte-expressed nicotinic acetylcholine receptors (nAChRs), α4β2 and α7. We recently found that Protein kinase B-β (Pkb-β or Akt2) expression is restricted to astrocytes in mice and humans. To determine if AKT2 plays a role in astrocytic nicotinic responses, we generated astrocyte-specific Akt2 conditional knockout (cKO) and full Akt2 KO mice for in vivo and in vitro experiments. For in vivo studies, we examined mice exposed to chronic nicotine for two weeks in drinking water (200 μg/mL) and following acute nicotine challenge (0.09, 0.2 mg/kg) after 24 hrs. Our in vitro studies used cultured mouse astrocytes to measure nicotine-dependent astrocytic responses. We validated our approaches using lipopolysaccharide (LPS) exposure inducing astrogliosis. Sholl analysis was used to measure glial fibrillary acidic protein responses in astrocytes. Our data show that wild-type (WT) mice exhibit increased astrocyte morphological complexity during acute nicotine exposure, with decreasing complexity during chronic nicotine use, whereas Akt2 cKO mice showed increased astrocyte morphology complexity. In culture, we found that 100μM nicotine was sufficient for morphological changes and blocking α7 or α4β2 nAChRs prevented observed morphologic changes. Finally, we performed conditioned place preference (CPP) in Akt2 cKO mice and found that astrocytic AKT2 deficiency reduced nicotine preference compared to controls. These findings show the importance of nAChRs and Akt2 signaling in the astrocytic response to nicotine.
Collapse
Affiliation(s)
- Andrew M. Lombardi
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
| | - Helen Wong
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Myra E. Bower
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Ryan Milstead
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Curtis Borski
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Emily Schmitt
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
| | - Mina Griffioen
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Lauren LaPlante
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Marissa A. Ehringer
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Jerry Stitzel
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Charles A. Hoeffer
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
- Linda Crnic Institute, Anschutz Medical Center, Aurora, CO 80045
| |
Collapse
|
3
|
Li D, Wang Y, Guo Y, Wang W. Bioinformatics analysis reveals multiple functional changes in astrocytes in temporal lobe epilepsy. Brain Res 2024; 1831:148820. [PMID: 38417653 DOI: 10.1016/j.brainres.2024.148820] [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: 06/08/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/01/2024]
Abstract
Epilepsy is a prevalent chronic neurological disorder characterized by recurrent seizures and brain dysfunction. Existing antiepileptic drugs (AEDs) mainly act on neurons and provide symptomatic control of seizures, but they do not modify the progression of epilepsy and may cause serious adverse effects. Increasing evidence suggests that reactive astrogliosis is critical in the pathophysiology of epilepsy. However, the function of reactive astrocytes in epilepsy has not been thoroughly explored. To provide a new perspective on the role of reactive astrocytes in epileptogenesis, we identified human astrocyte-specific genes and found 131 of these genes significantly differentially expressed in human temporal lobe epilepsy (TLE) datasets. Multiple astrocytic functions, such as cell adhesion, cell morphogenesis, actin filament-based process, apoptotic cell clearance and response to oxidative stress, were found to be promoted. Moreover, multiple altered astrocyte-specific genes were enriched in phagocytosis, perisynaptic astrocyte processes (PAPs), plasticity, and synaptic functions. Nine hub genes (ERBB2, GFAP, NOTCH2, ITGAV, ABCA1, AQP4, LRP1, GJA1, and YAP1) were identified by protein-protein interaction (PPI) network analysis. The correlation between the expression of these hub genes and seizure frequency, as well as epilepsy-related factors, including inflammatory mediators, complement factors, glutamate excitotoxicity and astrocyte reactivity, were analyzed. Additionally, upstream transcription factors of the hub genes were predicted. Finally, astrogliosis and the expression of the hub genes were validated in an epileptic rat model. Our findings reveal the various changes in astrocyte function associated with epilepsy and provide candidate astrocyte-specific genes that could be potential antiepileptogenic targets.
Collapse
Affiliation(s)
- Dongxiao Li
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei 050000, China; Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Yufeng Wang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yansu Guo
- Beijing Geriatric Healthcare Center, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Weiping Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei 050000, China; Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei 050000, China.
| |
Collapse
|
4
|
Lin W, Zhao XY, Cheng JW, Li LT, Jiang Q, Zhang YX, Han F. Signaling pathways in brain ischemia: Mechanisms and therapeutic implications. Pharmacol Ther 2023; 251:108541. [PMID: 37783348 DOI: 10.1016/j.pharmthera.2023.108541] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Ischemic stroke occurs when the arteries supplying blood to the brain are narrowed or blocked, inducing damage to brain tissue due to a lack of blood supply. One effective way to reduce brain damage and alleviate symptoms is to reopen blocked blood vessels in a timely manner and reduce neuronal damage. To achieve this, researchers have focused on identifying key cellular signaling pathways that can be targeted with drugs. These pathways include oxidative/nitrosative stress, excitatory amino acids and their receptors, inflammatory signaling molecules, metabolic pathways, ion channels, and other molecular events involved in stroke pathology. However, evidence suggests that solely focusing on protecting neurons may not yield satisfactory clinical results. Instead, researchers should consider the multifactorial and complex mechanisms underlying stroke pathology, including the interactions between different components of the neurovascular unit. Such an approach is more representative of the actual pathological process observed in clinical settings. This review summarizes recent research on the multiple molecular mechanisms and drug targets in ischemic stroke, as well as recent advances in novel therapeutic strategies. Finally, we discuss the challenges and future prospects of new strategies based on the biological characteristics of stroke.
Collapse
Affiliation(s)
- Wen Lin
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang-Yu Zhao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jia-Wen Cheng
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Li-Tao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Quan Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yi-Xuan Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China.
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China; Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
5
|
De Picker LJ, Morrens M, Branchi I, Haarman BCM, Terada T, Kang MS, Boche D, Tremblay ME, Leroy C, Bottlaender M, Ottoy J. TSPO PET brain inflammation imaging: A transdiagnostic systematic review and meta-analysis of 156 case-control studies. Brain Behav Immun 2023; 113:415-431. [PMID: 37543251 DOI: 10.1016/j.bbi.2023.07.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 06/26/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
INTRODUCTION The 18-kDa translocator protein (TSPO) is increasingly recognized as a molecular target for PET imaging of inflammatory responses in various central nervous system (CNS) disorders. However, the reported sensitivity and specificity of TSPO PET to identify brain inflammatory processes appears to vary greatly across disorders, disease stages, and applied quantification methods. To advance TSPO PET as a potential biomarker to evaluate brain inflammation and anti-inflammatory therapies, a better understanding of its applicability across disorders is needed. We conducted a transdiagnostic systematic review and meta-analysis of all in vivo human TSPO PET imaging case-control studies in the CNS. Specifically, we investigated the direction, strength, and heterogeneity associated with the TSPO PET signal across disorders in pre-specified brain regions, and explored the demographic and methodological sources of heterogeneity. METHODS We searched for English peer-reviewed articles that reported in vivo human case-control TSPO PET differences. We extracted the demographic details, TSPO PET outcomes, and technical variables of the PET procedure. A random-effects meta-analysis was applied to estimate case-control standardized mean differences (SMD) of the TSPO PET signal in the lobar/whole-brain cortical grey matter (cGM), thalamus, and cortico-limbic circuitry between different illness categories. Heterogeneity was evaluated with the I2 statistic and explored using subgroup and meta-regression analyses for radioligand generation, PET quantification method, age, sex, and publication year. Significance was set at the False Discovery Rate (FDR)-corrected P < 0.05. RESULTS 156 individual case-control studies were included in the systematic review, incorporating data for 2381 healthy controls and 2626 patients. 139 studies documented meta-analysable data and were grouped into 11 illness categories. Across all the illness categories, we observed a significantly higher TSPO PET signal in cases compared to controls for the cGM (n = 121 studies, SMD = 0.358, PFDR < 0.001, I2 = 68%), with a significant difference between the illness categories (P = 0.004). cGM increases were only significant for Alzheimer's disease (SMD = 0.693, PFDR < 0.001, I2 = 64%) and other neurodegenerative disorders (SMD = 0.929, PFDR < 0.001, I2 = 73%). Cortico-limbic increases (n = 97 studies, SMD = 0.541, P < 0.001, I2 = 67%) were most prominent for Alzheimer's disease, mild cognitive impairment, other neurodegenerative disorders, mood disorders and multiple sclerosis. Thalamic involvement (n = 79 studies, SMD = 0.393, P < 0.001, I2 = 71%) was observed for Alzheimer's disease, other neurodegenerative disorders, multiple sclerosis, and chronic pain and functional disorders (all PFDR < 0.05). Main outcomes for systemic immunological disorders, viral infections, substance use disorders, schizophrenia and traumatic brain injury were not significant. We identified multiple sources of between-study variance to the TSPO PET signal including a strong transdiagnostic effect of the quantification method (explaining 25% of between-study variance; VT-based SMD = 0.000 versus reference tissue-based studies SMD = 0.630; F = 20.49, df = 1;103, P < 0.001), patient age (9% of variance), and radioligand generation (5% of variance). CONCLUSION This study is the first overarching transdiagnostic meta-analysis of case-control TSPO PET findings in humans across several brain regions. We observed robust increases in the TSPO signal for specific types of disorders, which were widespread or focal depending on illness category. We also found a large and transdiagnostic horizontal (positive) shift of the effect estimates of reference tissue-based compared to VT-based studies. Our results can support future studies to optimize experimental design and power calculations, by taking into account the type of disorder, brain region-of-interest, radioligand, and quantification method.
Collapse
Affiliation(s)
- Livia J De Picker
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium.
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Roma, Italy
| | - Bartholomeus C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tatsuhiro Terada
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Min Su Kang
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, UK
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, BC, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Claire Leroy
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France
| | - Michel Bottlaender
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France; Université Paris-Saclay, UNIACT, Neurospin, CEA, Gif-sur-Yvette, France
| | - Julie Ottoy
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
6
|
Tripodi F, Motta Z, Murtas G, Rabattoni V, Nonnis S, Grassi Scalvini F, Rinaldi AM, Rizzi R, Bearzi C, Badone B, Sacchi S, Tedeschi G, Maffioli E, Coccetti P, Pollegioni L. Serine metabolism during differentiation of human iPSC-derived astrocytes. FEBS J 2023; 290:4440-4464. [PMID: 37166453 DOI: 10.1111/febs.16816] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/05/2023] [Accepted: 05/10/2023] [Indexed: 05/12/2023]
Abstract
Astrocytes are essential players in development and functions, being particularly relevant as regulators of brain energy metabolism, ionic homeostasis and synaptic transmission. They are also the major source of l-serine in the brain, which is synthesized from the glycolytic intermediate 3-phosphoglycerate through the phosphorylated pathway. l-Serine is the precursor of the two main co-agonists of the N-methyl-d-aspartate receptor, glycine and d-serine. Strikingly, dysfunctions in both l- and d-serine metabolism are associated with neurological and psychiatric disorders. Here, we exploited a differentiation protocol, based on the generation of human mature astrocytes from neural stem cells, and investigated the modification of the proteomic and metabolomic profile during the differentiation process. We show that differentiated astrocytes are more similar to mature rather than to reactive ones, and that axogenesis and pyrimidine metabolism increase up to 30 days along with the folate cycle and sphingolipid metabolism. Consistent with the proliferation and cellular maturation processes that are taking place, also the intracellular levels of l-serine, glycine, threonine, l- and d-aspartate (which level is unexpectedly higher than that of d-serine) show the same biosynthetic time course. A significant utilization of l-serine from the medium is apparent while glycine is first consumed and then released with a peak at 30 days, parallel to its intracellular level. These results underline how metabolism changes during astrocyte differentiation, highlight that d-serine synthesis is restricted in differentiated astrocytes and provide a valuable model for developing potential novel therapeutic approaches to address brain diseases, especially the ones related to serine metabolism alterations.
Collapse
Affiliation(s)
- Farida Tripodi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Italy
| | - Zoraide Motta
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Giulia Murtas
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Valentina Rabattoni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Simona Nonnis
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, Italy
| | | | | | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy
- Department of Medical-Surgical Science and Biotechnologies, University of Rome La Sapienza, Italy
| | - Claudia Bearzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy
- Institute for Biomedical Technologies, National Research Council of Italy (ITB-CNR), Milan, Italy
| | - Beatrice Badone
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Italy
| | - Silvia Sacchi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Gabriella Tedeschi
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, Italy
- CIMAINA, University of Milano, Italy
| | - Elisa Maffioli
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, Italy
| | - Paola Coccetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
7
|
Zhang W, Wang T, Xue Y, Zhan B, Lai Z, Huang W, Peng X, Zhou Y. Research progress of extracellular vesicles and exosomes derived from mesenchymal stem cells in the treatment of oxidative stress-related diseases. Front Immunol 2023; 14:1238789. [PMID: 37646039 PMCID: PMC10461809 DOI: 10.3389/fimmu.2023.1238789] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
There is growing evidence that mesenchymal stem cell-derived extracellular vesicles and exosomes can significantly improve the curative effect of oxidative stress-related diseases. Mesenchymal stem cell extracellular vesicles and exosomes (MSC-EVs and MSC-Exos) are rich in bioactive molecules and have many biological regulatory functions. In this review, we describe how MSC-EVs and MSC-Exos reduce the related markers of oxidative stress and inflammation in various systemic diseases, and the molecular mechanism of MSC-EVs and MSC-Exos in treating apoptosis and vascular injury induced by oxidative stress. The results of a large number of experimental studies have shown that both local and systemic administration can effectively inhibit the oxidative stress response in diseases and promote the survival and regeneration of damaged parenchymal cells. The mRNA and miRNAs in MSC-EVs and MSC-Exos are the most important bioactive molecules in disease treatment, which can inhibit the apoptosis, necrosis and oxidative stress of lung, heart, kidney, liver, bone, skin and other cells, and promote their survive and regenerate.
Collapse
Affiliation(s)
- Wenwen Zhang
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Tingyu Wang
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yuanye Xue
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Bingbing Zhan
- School of Pharmaceutical Sciences, Guangdong Medical University, Dongguan, China
| | - Zengjie Lai
- The Second Clinical Medical College of Guangdong Medical University, Dongguan, China
| | - Wenjie Huang
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xinsheng Peng
- Biomedical Innovation Center, Guangdong Medical University, Dongguan, China
- Institute of Marine Medicine, Guangdong Medical University, Zhanjiang, China
| | - Yanfang Zhou
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
8
|
Savtchenko LP, Rusakov DA. Glutamate-Transporter Unbinding in Probabilistic Synaptic Environment Facilitates Activation of Distant NMDA Receptors. Cells 2023; 12:1610. [PMID: 37371080 DOI: 10.3390/cells12121610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Once outside the synaptic cleft, the excitatory neurotransmitter glutamate is rapidly bound by its high-affinity transporters, which are expressed in abundance on the surface of perisynaptic astroglia. While this binding and the subsequent uptake of glutamate constrain excitatory transmission mainly within individual synapses, there is growing evidence for the physiologically important extrasynaptic actions of glutamate. However, the mechanistic explanation and the scope of such actions remain obscure. Furthermore, a significant proportion of glutamate molecules initially bound by transporters could be released back into the extracellular space before being translocated into astrocytes. To understand the implications of such effects, we simulated the release, diffusion, and transporter and receptor interactions of glutamate molecules in the synaptic environment. The latter was represented via trial-by-trial stochastic generation of astroglial and neuronal elements in the brain neuropil (overlapping spheroids of varied sizes), rather than using the 'average' morphology, thus reflecting the probabilistic nature of neuropil architectonics. Our simulations predict significant activation of high-affinity receptors, such as receptors of the NMDA type, at distances beyond half-micron from the glutamate release site, with glutamate-transporter unbinding playing an important role. These theoretical predictions are consistent with recent glutamate imaging data, thus lending support to the concept of significant volume-transmitted actions of glutamate in the brain.
Collapse
Affiliation(s)
- Leonid P Savtchenko
- UCL Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
9
|
Astrocytes in the pathophysiology of neuroinfection. Essays Biochem 2023; 67:131-145. [PMID: 36562155 DOI: 10.1042/ebc20220082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Key homeostasis providing cells in the central nervous system (CNS) are astrocytes, which belong to the class of cells known as atroglia, a highly heterogeneous type of neuroglia and a prominent element of the brain defence. Diseases evolve due to altered homeostatic state, associated with pathology-induced astroglia remodelling represented by reactive astrocytes, astroglial atrophy and astrodegeneration. These features are hallmarks of most infectious insults, mediated by bacteria, protozoa and viruses; they are also prominent in the systemic infection. The COVID-19 pandemic revived the focus into neurotropic viruses such as SARS-CoV2 (Coronaviridae) but also the Flaviviridae viruses including tick-borne encephalitis (TBEV) and Zika virus (ZIKV) causing the epidemic in South America prior to COVID-19. Astrocytes provide a key response to neurotropic infections in the CNS. Astrocytes form a parenchymal part of the blood-brain barrier, the site of virus entry into the CNS. Astrocytes exhibit aerobic glycolysis, a form of metabolism characteristic of highly morphologically plastic cells, like cancer cells, hence a suitable milieu for multiplication of infectious agent, including viral particles. However, why the protection afforded by astrocytes fails in some circumstances is an open question to be studied in the future.
Collapse
|
10
|
Vasciaveo V, Iadarola A, Casile A, Dante D, Morello G, Minotta L, Tamagno E, Cicolin A, Guglielmotto M. Sleep fragmentation affects glymphatic system through the different expression of AQP4 in wild type and 5xFAD mouse models. Acta Neuropathol Commun 2023; 11:16. [PMID: 36653878 PMCID: PMC9850555 DOI: 10.1186/s40478-022-01498-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/18/2022] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by genetic and multifactorial risk factors. Many studies correlate AD to sleep disorders. In this study, we performed and validated a mouse model of AD and sleep fragmentation, which properly mimics a real condition of intermittent awakening. We noticed that sleep fragmentation induces a general acceleration of AD progression in 5xFAD mice, while in wild type mice it affects cognitive behaviors in particular learning and memory. Both these events may be correlated to aquaporin-4 (AQP4) modulation, a crucial player of the glymphatic system activity. In particular, sleep fragmentation differentially affects aquaporin-4 channel (AQP4) expression according to the stage of the disease, with an up-regulation in younger animals, while such change cannot be detected in older ones. Moreover, in wild type mice sleep fragmentation affects cognitive behaviors, in particular learning and memory, by compromising the glymphatic system through the decrease of AQP4. Nevertheless, an in-depth study is needed to better understand the mechanism by which AQP4 is modulated and whether it could be considered a risk factor for the disease development in wild type mice. If our hypotheses are going to be confirmed, AQP4 modulation may represent the convergence point between AD and sleep disorder pathogenic mechanisms.
Collapse
Affiliation(s)
- Valeria Vasciaveo
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy ,grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Antonella Iadarola
- grid.432329.d0000 0004 1789 4477Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza, Corso Bramante 88, 10126 Turin, Italy
| | - Antonino Casile
- grid.5602.10000 0000 9745 6549School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 9, 62032 Camerino, MC Italy
| | - Davide Dante
- grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Giulia Morello
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy ,grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Lorenzo Minotta
- grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Elena Tamagno
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy ,grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Alessandro Cicolin
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy
| | - Michela Guglielmotto
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy ,grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| |
Collapse
|
11
|
Liu BP, Zhang C, Zhang YP, Li KW, Song C. The combination of chronic stress and smoke exacerbated depression-like changes and lung cancer factor expression in A/J mice: Involve inflammation and BDNF dysfunction. PLoS One 2022; 17:e0277945. [PMID: 36417428 PMCID: PMC9683596 DOI: 10.1371/journal.pone.0277945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Depression is positively correlated with the high incidence and low survival rate of cancers, while more cancer patients suffer depression. However, the interaction between depression and cancer, and possible underline mechanisms are unclear. METHODS Chronic unpredictable mild stress (CUMS) was used to induce depression, and smoke to induce lung cancer in lung cancer vulnerable AJ mice. After 8 weeks, sucrose preference and forced swimming behaviors were tested. Blood corticosterone concentration, and levels of cytokines, lung cancer-related factors, brain-derived neurotrophic factor (BDNF) and apoptosis-related factors in the lung, amygdala and hippocampus were measured. RESULTS Compared to control group, CUMS or smoke decreased sucrose consumption and increased immobility time, which were deteriorated by stress+smoke. CUMS, smoke or both combination decreased mononuclear viability and lung TNF-α concentration, increased serum corticosterone and lung interleukin (IL)-1, IL-2, IL-6, IL-8, IL-10, IL-12 and HSP-90α concentrations. Furthermore, stress+smoke caused more increase in corticosterone and IL-10, but decreased TNF-α. In parallel, in the lung, Bcl-2/Bax and lung cancer-related factors CDK1, CDC20, P38α etc were significantly increased in stress+smoke group. Moreover, CUMS decreased BDNF, while CUMS or smoke increased TrkB and P75 concentrations, which were exacerbated by stress+smoke. In the amygdala, except for CUMS largely increased Bax/Bcl-2 and decreased TrkB, each single factor decreased BDNF and IL-10, but increased P75, IL-1β, IL-12, TNF-α concentrations. Changes in Bax/Bcl-2, IL-10 and TNF-α were further aggravated by the combination. In the hippocampus, except for CUMS largely increased P75 concentration, each single factor significantly increased Bax/Bcl-2 ratio, IL-1β and TNF-α, but decreased BDNF, TrkB and IL-10 concentrations. Changes in Bax, Bax/Bcl-2, IL-10 and TNF-α were further aggravated by the combination. CONCLUSION These results suggest that a synergy between CUMS and smoke exposure could promote the development of depression and lung cancer, through CUMS increased the risk of cancer occurrence, and conversely lung cancer inducer smoke exposure deteriorated depressive symptoms.
Collapse
Affiliation(s)
- Bai-Ping Liu
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
- Key laboratory of Aquatic Product Processing, Guangdong Ocean University, Zhanjiang, China
| | - Cai Zhang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
- Key laboratory of Aquatic Product Processing, Guangdong Ocean University, Zhanjiang, China
| | - Yong-Ping Zhang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
- Key laboratory of Aquatic Product Processing, Guangdong Ocean University, Zhanjiang, China
- Marine Medical Research and Development Centre, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
| | - Kang-Wei Li
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
- Key laboratory of Aquatic Product Processing, Guangdong Ocean University, Zhanjiang, China
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
- Key laboratory of Aquatic Product Processing, Guangdong Ocean University, Zhanjiang, China
- Marine Medical Research and Development Centre, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- * E-mail:
| |
Collapse
|
12
|
Zhou L, Liang J, Xiong T. Research progress of mesenchymal stem cell-derived exosomes on inflammatory response after ischemic stroke. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:500-506. [PMID: 37202091 PMCID: PMC10264999 DOI: 10.3724/zdxbyxb-2022-0077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/24/2022] [Indexed: 05/20/2023]
Abstract
Ischemic stroke is characterized by cute onset and high mortality. The suppression of neuroinflammation is crucial in the treatment of ischemic stroke. Exosomes derived from mesenchymal stem cell (MSC) have attracted extensive research attention due to their wide origin, small size, and containing large number of active components. Recent studies have shown that MSC-derived exosomes can inhibit the proinflammatory activity of microglia and astrocytes and stimulate their neuroprotective activity; also can inhibit neuroinflammation by regulating immune cells and inflammatory mediators. This article reviews the roles and related mechanism of MSC-derived exosomes in neuroinflammation after ischemic stroke, hoping to provide ideas and references for the development of a novel approach for the treatment of ischemic stroke diseases.
Collapse
Affiliation(s)
- Lujia Zhou
- 1. Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Jingyan Liang
- 1. Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu Province, China
- 2. Jiangsu Provincial Key Laboratory of Geriatric Disease Prevention and Control, Yangzhou 225001, Jiangsu Province, China
| | - Tianqing Xiong
- 1. Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu Province, China
- 2. Jiangsu Provincial Key Laboratory of Geriatric Disease Prevention and Control, Yangzhou 225001, Jiangsu Province, China
| |
Collapse
|
13
|
Sun X, Li X, Zhou Y, Wang Y, Liu X. Exogenous TIPE2 Inhibit TAK1 to Improve Inflammation and Neuropathic Pain Induced by Sciatic Nerve Injury Through Inactivating NF-κB and JNK. Neurochem Res 2022; 47:3167-3177. [PMID: 35842555 PMCID: PMC9470725 DOI: 10.1007/s11064-022-03671-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022]
Abstract
Tumor necrosis factor-alpha-induced protein 8-like 2 (TIPE2) possesses potent anti-inflammatory effect. However, if TIPE2 ameliorates sciatic nerve injury (SNI)-induced inflammation and pain remains undiscussed, and the underlying role TAK1 in it were unknown. To verify our imagine, we performed SNI surgery, and analyzed expression and colocalization of TIPE2 and TAK1 in spinal cord and dorsal root neurons (DRG) by immunofluorescence staining and western blot. And the biological analysis, inflammatory factors, and pathological improvement were determined, and the regulation of TIPE2 in TAK1, phosphor-NF-κB, phospho-JNK was also tested by immunofluorescence staining and western blot. Experimental results showed the parabola-like change of TIPE2 and rising expression of TAK1 in spinal cord and DRG. And intrathecal TIPE2 injection could significantly improve the status of SNI rats, inhibit level of IL-6, IL-10 and TNF-α, raise the thermal withdrawal relax latency and mechanical withdrawal thresholds. Meanwhile, we also detected how TIPE2 regulated TAK1, and the downstream pathway NF-κB and JNK. The result indicated that TIPE2 could reduce TAK1 expression, and make NF-κB and JNK inactivated. To deeply discuss the potential mechanism, we injected TAK1 oligodeoxynucleotide into rats, and found that TIPE2 exerted the protective role against SNI through TAK1. In brief, TIPE2 reduced expression of TAK1, thereby inhibiting activation of NF-kB and JNK, further improving the neuroinflammation and neuropathic pain. TIPE2 played a protective role in sciatic nerve injury rats through regulating TAK1.
Collapse
Affiliation(s)
- Xuehua Sun
- Pain department, Yantai Affiliated Hospital of Binzhou Medical University, No. 717, Jinbu street, Muping District, Yantai City, 264100, Shandong, People's Republic of China.
| | - Xinyou Li
- Pain department, Yantai Affiliated Hospital of Binzhou Medical University, No. 717, Jinbu street, Muping District, Yantai City, 264100, Shandong, People's Republic of China
| | - Youfei Zhou
- Pain department, Yantai Affiliated Hospital of Binzhou Medical University, No. 717, Jinbu street, Muping District, Yantai City, 264100, Shandong, People's Republic of China
| | - Yufei Wang
- Pain department, Yantai Affiliated Hospital of Binzhou Medical University, No. 717, Jinbu street, Muping District, Yantai City, 264100, Shandong, People's Republic of China
| | - Xiaochen Liu
- Pain department, Yantai Affiliated Hospital of Binzhou Medical University, No. 717, Jinbu street, Muping District, Yantai City, 264100, Shandong, People's Republic of China
| |
Collapse
|
14
|
Xu GY, Xu S, Zhang YX, Yu ZY, Zou F, Ma XS, Xia XL, Zhang WJ, Jiang JY, Song J. Cell-Free Extracts from Human Fat Tissue with a Hyaluronan-Based Hydrogel Attenuate Inflammation in a Spinal Cord Injury Model through M2 Microglia/Microphage Polarization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107838. [PMID: 35333441 DOI: 10.1002/smll.202107838] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/25/2022] [Indexed: 06/14/2023]
Abstract
Treatment for spinal cord injuries (SCIs) is often ineffective because SCIs result in a loss of nerve tissue, glial scar formation, local ischemia and secondary inflammation. The current promising strategy for SCI is the combination of bioactive materials and cytokines. Bioactive materials support the injured spinal cord, stabilize the morphology, and avoid excessive inflammatory responses. Fat extract (FE) is a cell-free liquid component containing a variety of cytokines extracted from human fat tissue using mechanical methods. In this research, a biocompatible HAMC (hyaluronan and methylcellulose) loaded with FE is used to treat a model of spinal cord contusion in mice. The composite not only inhibits death of neuro- and vascular cells and leads to the preservation of neural and vascular structure, but also modulates the inflammatory phenotype of macrophages in the locally injured region. Specifically, FE promotes the polarization of macrophages from an inflammatory M1 phenotype to an anti-inflammatory M2 phenotype. During the screening of the involved pathways, it is corroborated that activation of the STAT6/Arg-1 signaling pathway is involved in macrophage M2 polarization. In summary, FE is a promising treatment for SCI, as it is easy to obtain, nonimmunogenic, and effective.
Collapse
Affiliation(s)
- Guang-Yu Xu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shun Xu
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Yu-Xuan Zhang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zi-You Yu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200011, China
| | - Fei Zou
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiao-Sheng Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xin-Lei Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wen-Jie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200011, China
| | - Jian-Yuan Jiang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jian Song
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
15
|
Soares-Silva B, Beserra-Filho JIA, Morera PMA, Custódio-Silva AC, Maria-Macêdo A, Silva-Martins S, Alexandre-Silva V, Silva SP, Silva RH, Ribeiro AM. The bee venom active compound melittin protects against bicuculline-induced seizures and hippocampal astrocyte activation in rats. Neuropeptides 2022; 91:102209. [PMID: 34808488 DOI: 10.1016/j.npep.2021.102209] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/26/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022]
Abstract
Epilepsy is a chronic neuropathology characterized by an abnormal hyperactivity of neurons that generate recurrent, spontaneous, paradoxical and synchronized nerve impulses, leading or not to seizures. This neurological disorder affects around 70 million individuals worldwide. Pharmacoresistance is observed in about 30% of the patients and long-term use of antiepileptics may induce serious side effects. Thus, there is an interest in the study of the therapeutic potential of bioactive substances isolated from natural products in the treatment of epilepsy. Arthropod venoms contain neurotoxins that have high affinity for molecular structures in the neural tissue such as receptors, transporters and ion channels both in glial and neuronal membranes. This study evaluated the potential neuroprotective effect of melittin (MEL), an active compound of bee venom, in the bicuculline-induced seizure model (BIC) in rats. Male Wistar rats (3 months, 250-300 g) were submitted to surgery for the implantation of a unilateral cannula in the lateral ventricle. After the recovery period, rats received a microinjection of saline solution or MEL (0.1 mg per animal). Firstly, rats were evaluated in the open field (20 min) and in the elevated plus maze (5 min) tests after received microinjection of saline or MEL. After, 30 min later animals received BIC (100 mg/ml) or saline, and their behaviors were analyzed for 20 min in the open field according to a seizure scale. At the end, rats were euthanized, brains collected and processed to glial fibrillary acidic protein (GFAP) immunohistochemistry evaluation. No changes were observed in MEL-treated rats in the open field and elevated plus maze. However, 90% of MEL-treated animals were protected against seizures induced by BIC. There was an increase in the latency for the onset of seizures, accompanied by a reduction of GFAP-immunoreactivity cells in the dentate gyrus and CA1. Thus, our study suggests that MEL has an anticonvulsant potential, and further studies are needed to elucidate the mechanisms involved in this action.
Collapse
Affiliation(s)
| | - José Ivo Araújo Beserra-Filho
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, Brazil; Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Amanda Maria-Macêdo
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, Brazil
| | | | | | - Sara Pereira Silva
- Departament of Biosciences, Universidade Federal de São Paulo, Santos, Brazil
| | - Regina Helena Silva
- Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
16
|
Balachandar L, Borrego D, Diaz JR. Serotype-based evaluation of an optogenetic construct in rat cortical astrocytes. Biochem Biophys Res Commun 2022; 593:35-39. [DOI: 10.1016/j.bbrc.2022.01.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/08/2022] [Indexed: 11/16/2022]
|
17
|
Ahdoot-Levi H, Croitoru O, Bareli T, Sudai E, Peér-Nissan H, Jacob A, Gispan I, Maayan R, Weizman A, Yadid G. The Effect of Dehydroepiandrosterone Treatment on Neurogenesis, Astrogliosis and Long-Term Cocaine-Seeking Behavior in a Cocaine Self-Administration Model in Rats. Front Neurosci 2021; 15:773197. [PMID: 34899172 PMCID: PMC8662380 DOI: 10.3389/fnins.2021.773197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Cocaine addiction is an acquired behavioral state developed in vulnerable individuals after cocaine exposure. It is characterized by compulsive drug-seeking and high vulnerability to relapse even after prolonged abstinence, associated with decreased neurogenesis in the hippocampus. This addictive state is hypothesized to be a form of “memory disease” in which the drug exploits the physiological neuroplasticity mechanisms that mediate regular learning and memory processes. Therefore, a major focus of the field has been to identify the cocaine-induced neuroadaptations occurring in the usurped brain’s reward circuit. The neurosteroid dehydroepiandrosterone (DHEA) affects brain cell morphology, differentiation, neurotransmission, and memory. It also reduces drug-seeking behavior in an animal model of cocaine self-administration. Here, we examined the long-lasting effects of DHEA treatment on the attenuation of cocaine-seeking behavior. We also examined its short- and long-term influence on hippocampal cells architecture (neurons and astrocytes). Using a behavioral examination, immunohistochemical staining, and diffusion tensor imaging, we found an immediate effect on tissue density and activation of astrocytes, which has a continuous beneficial effect on neurogenesis and tissue organization. This research emphasizes the requites concert between astrocytes and neurons in the rehabilitation from addiction behavior. Thus, DHEA may serve as a treatment that corrects brain damage following exposure to and abstinence from cocaine.
Collapse
Affiliation(s)
- Hadas Ahdoot-Levi
- Neuropharmacology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Ramat-Gan, Israel
| | - Ofri Croitoru
- Neuropharmacology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Ramat-Gan, Israel
| | - Tzofnat Bareli
- Neuropharmacology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Ramat-Gan, Israel
| | - Einav Sudai
- Neuropharmacology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Ramat-Gan, Israel.,The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Ramat-Gan, Israel
| | - Hilla Peér-Nissan
- Neuropharmacology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Ramat-Gan, Israel
| | - Avi Jacob
- Neuropharmacology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Ramat-Gan, Israel
| | - Iris Gispan
- Neuropharmacology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Ramat-Gan, Israel
| | - Rachel Maayan
- The Laboratory of Biological Psychiatry, Felsenstein Medical Research Center and Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Abraham Weizman
- The Laboratory of Biological Psychiatry, Felsenstein Medical Research Center and Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel.,Research Unit, Geha Mental Health Center, Petah Tikva, Israel
| | - Gal Yadid
- Neuropharmacology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Ramat-Gan, Israel.,The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Ramat-Gan, Israel
| |
Collapse
|
18
|
Verkhratsky A, Li B, Scuderi C, Parpura V. Principles of Astrogliopathology. ADVANCES IN NEUROBIOLOGY 2021; 26:55-73. [PMID: 34888830 DOI: 10.1007/978-3-030-77375-5_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The role of astrocytes in the nervous system pathology was early on embraced by neuroscientists at end of the nineteenth and the beginning of the twentieth century, only to be pushed aside by neurone-centric dogmas during most of the twentieth century. However, the last decade of the twentieth century and the twenty-first century have brought the astroglial "renaissance", which has put astroglial cells as key players in pathophysiology of most if not all disorders of the nervous system and has regarded astroglia as a fertile ground for therapeutic intervention.Astrocytic contribution to neuropathology can be primary, whereby cell-autonomous changes, such as mutations in gene encoding for glial fibrillary acidic protein, can drive the pathologic progression, in this example, Alexander disease. They can also be secondary, when astrocytes respond to a variety of insults to the nervous tissue. Regardless of their origin, being cell-autonomous or not, changes in astroglia that occur in pathology, that is, astrogliopathology, can be contemporary and arbitrary classified into four forms: (i) reactive astrogliosis, (ii) astrocytic atrophy with loss of function, (iii) pathological remodelling of astrocytes and (iv) astrodegeneration morphologically manifested as clasmatodendrosis. Inevitably, as with any other classification, this classification of astrogliopathology awaits its revision that shall be rooted in new discoveries and concepts.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Baoman Li
- Practical Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
19
|
Kubrusly RCC, da Rosa Valli T, Ferreira MNMR, de Moura P, Borges-Martins VPP, Martins RS, Ferreira DDP, Sathler MF, de Melo Reis RA, Ferreira GC, Manhães AC, Dos Santos Pereira M. Caffeine Improves GABA Transport in the Striatum of Spontaneously Hypertensive Rats (SHR). Neurotox Res 2021; 39:1946-1958. [PMID: 34637050 DOI: 10.1007/s12640-021-00423-0] [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: 03/09/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022]
Abstract
The spontaneously hypertensive rat (SHR) is an excellent animal model that mimics the behavioral and neurochemical phenotype of attention-deficit/hyperactivity disorder (ADHD). Here, we characterized the striatal GABA transport of SHR and investigated whether caffeine, a non-selective antagonist of adenosine receptors, could influence GABAergic circuitry. For this purpose, ex vivo striatal slices of SHR and Wistar (control strain) on the 35th postnatal day were dissected and incubated with [3H]-GABA to quantify the basal levels of uptake and release. SHR exhibited a reduced [3H]-GABA uptake and release, suggesting a defective striatal GABAergic transport system. GAT-1 appears to be the primary transporter for [3H]-GABA uptake in SHR striatum, as GAT-1 selective blocker, NO-711, completely abolished it. We also verified that acute exposure of striatal slices to caffeine improved [3H]-GABA uptake and release in SHR, whereas Wistar rats were not affected. GABA-uptake increase and cAMP accumulation promoted by caffeine was reverted by A1R activation with N6-cyclohexyl adenosine (CHA). As expected, the pharmacological blockade of cAMP-PKA signaling by H-89 also prevented caffeine-mediated [3H]-GABA uptake increment. Interestingly, a single caffeine exposure did not affect GAT-1 or A1R protein density in SHR, which was not different from Wistar protein levels, suggesting that the GAT-1-dependent transport in SHR has a defective functional activity rather than lower protein expression. The current data support that caffeine regulates GAT-1 function and improves striatal GABA transport via A1R-cAMP-PKA signaling, specifically in SHR. These results reinforce that caffeine may have therapeutic use in disorders where the GABA transport system is impaired.
Collapse
Affiliation(s)
| | | | | | - Pâmella de Moura
- Laboratório de Neurofarmacologia, Instituto Biomédico, Niterói, RJ, Brazil
| | | | - Robertta Silva Martins
- Laboratório de Neurofarmacologia, Instituto Biomédico, Niterói, RJ, Brazil
- Laboratório de Neurobiologia Celular E Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Ricardo Augusto de Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Costa Ferreira
- Laboratório de Neuroenergética E Erros Inatos Do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alex Christian Manhães
- Laboratório de Neurofisiologia, Instituto de Biologia, Universidade Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maurício Dos Santos Pereira
- Laboratório de Neurofarmacologia, Instituto Biomédico, Niterói, RJ, Brazil.
- Laboratório de Neurofisiologia Molecular, Departamento de Biologia Básica E Oral, Faculdade de Odontologia de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
20
|
Lalo U, Koh W, Lee CJ, Pankratov Y. The tripartite glutamatergic synapse. Neuropharmacology 2021; 199:108758. [PMID: 34433089 DOI: 10.1016/j.neuropharm.2021.108758] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/25/2021] [Accepted: 08/20/2021] [Indexed: 12/31/2022]
Abstract
Astroglial cells were long considered as structural and metabolic supporting cells are which do not directly participate in information processing in the brain. Discoveries of responsiveness of astrocytes to synaptically-released glutamate and their capability to release agonists of glutamate receptors awakened extensive studies of glia-neuron communications and led to the revolutionary changes in our understanding of brain cellular networks. Nowadays, astrocytes are widely acknowledged as inseparable element of glutamatergic synapses and role for glutamatergic astrocyte-neuron interactions in the brain computation is emerging. Astroglial glutamate receptors, in particular of NMDA, mGluR3 and mGluR5 types, can activate a variety of molecular cascades leading astroglial-driven modulation of extracellular levels of glutamate and activity of neuronal glutamate receptors. Their preferential location to the astroglial perisynaptic processes facilitates interaction of astrocytes with individual excitatory synapses. Bi-directional glutamatergic communication between astrocytes and neurons underpins a complex, spatially-distributed modulation of synaptic signalling thus contributing to the enrichment of information processing by the neuronal networks. Still, further research is needed to bridge the substantial gaps in our understanding of mechanisms and physiological relevance of astrocyte-neuron glutamatergic interactions, in particular ability of astrocytes directly activate neuronal glutamate receptors by releasing glutamate and, arguably, d-Serine. An emerging roles for aberrant changes in glutamatergic astroglial signalling, both neuroprotective and pathogenic, in neurological and neurodegenerative diseases also require further investigation. This article is part of the special Issue on 'Glutamate Receptors - The Glutamatergic Synapse'.
Collapse
Affiliation(s)
- Ulyana Lalo
- School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, South Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, South Korea
| | - Yuriy Pankratov
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| |
Collapse
|
21
|
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
|
22
|
Martins-Macedo J, Salgado AJ, Gomes ED, Pinto L. Adult brain cytogenesis in the context of mood disorders: From neurogenesis to the emergent role of gliogenesis. Neurosci Biobehav Rev 2021; 131:411-428. [PMID: 34555383 DOI: 10.1016/j.neubiorev.2021.09.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/06/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022]
Abstract
Psychiatric disorders severely impact patients' lives. Motivational, cognitive and emotional deficits are the most common symptoms observed in these patients and no effective treatment is still available, either due to the adverse side effects or the low rate of efficacy of currently available drugs. Neurogenesis recovery has been one important focus in the treatment of psychiatric disorders, which undeniably contributes to the therapeutic action of antidepressants. However, glial plasticity is emerging as a new strategy to explore the deficits observed in mood disorders and the efficacy of therapeutic interventions. Thus, it is crucial to understand the mechanisms behind glio- and neurogenesis to better define treatments and preventive therapies, once adult cytogenesis is of pivotal importance to cognitive and emotional components of behavior, both in healthy and pathological contexts, including in psychiatric disorders. Here, we review the concepts and history of neuro- and gliogenesis, providing as well a reflection on the functional importance of cytogenesis in the context of disease.
Collapse
Affiliation(s)
- Joana Martins-Macedo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
23
|
Virmani G, D'almeida P, Nandi A, Marathe S. Subfield-specific effects of chronic mild unpredictable stress on hippocampal astrocytes. Eur J Neurosci 2021; 54:5730-5746. [PMID: 33866634 DOI: 10.1111/ejn.15234] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/04/2021] [Indexed: 12/20/2022]
Abstract
Major depressive disorder (MDD) is a debilitating neuropsychiatric illness affecting over 20% of the population worldwide. Despite its prevalence, our understanding of its pathophysiology is severely limited, thus hampering the development of novel therapeutic strategies. Recent advances have clearly established astrocytes as major players in the pathophysiology, and plausibly pathogenesis, of major depression. In particular, astrocyte density in the hippocampus is severely diminished in MDD patients and correlates strongly with the disease outcome. Moreover, astrocyte densities from different subfields of the hippocampus show varying trends in terms of their correlation to the disease outcome. Given the central role that hippocampus plays in the pathophysiology of depression and in the action of antidepressant drugs, changes in hippocampal astrocyte density and physiology may have a significant effect on behavioral symptoms of MDD. In this study, we used chronic mild unpredictable stress (CMUS) in mice, which induces a depressive-like state, and examined its effects on astrocytes from different subfields of the hippocampus. We used SOX9 and S100β immunostaining to estimate the number of astrocytes per square millimeter from various hippocampal subfields. Furthermore, using confocal images of fluorescently labeled glial fibrillary acidic protein (GFAP)-immunopositive hippocampal astrocytes, we quantified various morphology-related parameters and performed Sholl analysis. We found that CMUS exerts differential effects on astrocyte cell numbers, ramification, cell radius, surface area, and process width of hippocampal astrocytes from different hippocampal subfields. Taken together, our study reveals that chronic stress does not uniformly affect all hippocampal astrocytes; but exerts its effects differentially on different astrocytic subpopulations within the hippocampus.
Collapse
Affiliation(s)
- Garima Virmani
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Priyal D'almeida
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Arnab Nandi
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Swananda Marathe
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| |
Collapse
|
24
|
Ramos-Gonzalez P, Mato S, Chara JC, Verkhratsky A, Matute C, Cavaliere F. Astrocytic atrophy as a pathological feature of Parkinson's disease with LRRK2 mutation. NPJ Parkinsons Dis 2021; 7:31. [PMID: 33785762 PMCID: PMC8009947 DOI: 10.1038/s41531-021-00175-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/03/2021] [Indexed: 02/01/2023] Open
Abstract
The principal hallmark of Parkinson's disease (PD) is the selective neurodegeneration of dopaminergic neurones. Mounting evidence suggests that astrocytes may contribute to dopaminergic neurodegeneration through decreased homoeostatic support and deficient neuroprotection. In this study, we generated induced pluripotent stem cells (iPSC)-derived astrocytes from PD patients with LRRK2(G2019S) mutation and healthy donors of the similar age. In cell lines derived from PD patients, astrocytes were characterised by a significant decrease in S100B and GFAP-positive astrocytic profiles associated with marked decrease in astrocyte complexity. In addition, PD-derived astrocytes demonstrated aberrant mitochondrial morphology, decreased mitochondrial activity and ATP production along with an increase of glycolysis and increased production of reactive oxygen species. Taken together, our data indicate that astrocytic asthenia observed in patient-derived cultures with LRRK2(G2019S) mutation may contribute to neuronal death through decreased homoeostatic support, elevated oxidative stress and failed neuroprotection.
Collapse
Affiliation(s)
- Paula Ramos-Gonzalez
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Susana Mato
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Biocruces, Bizkaia, Barakaldo, Spain
| | - Juan Carlos Chara
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alexei Verkhratsky
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Fabio Cavaliere
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain.
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
25
|
Tetragonia tetragonioides Relieves Depressive-Like Behavior through the Restoration of Glial Loss in the Prefrontal Cortex. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8888841. [PMID: 33628324 PMCID: PMC7895589 DOI: 10.1155/2021/8888841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/08/2021] [Accepted: 01/29/2021] [Indexed: 11/18/2022]
Abstract
Tetragonia tetragonioides, which is a halophyte and grows widely in Asian-Pacific regions, has been used for the treatment of digestive disorders in traditional oriental medicine. This study examined the potential antidepressant effect of Tetragonia tetragonioides in an astroglial degeneration model of depression, which was established based on the postmortem study of depressive patients' brain presenting diminished astrocytes in the prefrontal cortex. C57BL/6 male mice were exposed to glial ablation in the prefrontal cortex by the administration of the gliotoxin, L-alpha-aminoadipic acid (L-AAA) to induce depression. Tetragonia tetragonioides at doses of 100 mg/kg and 300 mg/kg, imipramine at a dose of 15 mg/kg, and distilled water were orally administrated to mice for 18 days. Behavioral tests including the open field test (OFT), sucrose preference test (SPT), forced swimming test (FST), and tail suspension test (TST) were carried out after 2 days of L-AAA injection. The expression levels of GFAP and NeuN in the prefrontal cortex were determined by immunohistochemistry. Mice subjected to glial ablation in the prefrontal cortex displayed decreased sucrose consumption in SPT and increased immobility time in FST and TST. Treatment with imipramine and Tetragonia tetragonioides remarkably ameliorated the behavioral despair induced by L-AAA. In addition, immunohistochemistry analysis showed that treatment with Tetragonia tetragonioides significantly restored the glial loss as indicated by the elevated GFAP expression level. These findings suggest that Tetragonia tetragonioides exerts an antidepressant effect through the restoration of glial loss under conditions of depression and can be a candidate for an antidepressant agent.
Collapse
|
26
|
Ceria nanoparticles ameliorate white matter injury after intracerebral hemorrhage: microglia-astrocyte involvement in remyelination. J Neuroinflammation 2021; 18:43. [PMID: 33588866 PMCID: PMC7883579 DOI: 10.1186/s12974-021-02101-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) can induce excessive accumulation of reactive oxygen species (ROS) that may subsequently cause severe white matter injury. The process of oligodendrocyte progenitor cell (OPC) differentiation is orchestrated by microglia and astrocytes, and ROS also drives the activation of microglia and astrocytes. In light of the potent ROS scavenging capacity of ceria nanoparticles (CeNP), we aimed to investigate whether treatment with CeNP ameliorates white matter injury by modulating ROS-induced microglial polarization and astrocyte alteration. Methods ICH was induced in vivo by collagenase VII injection. Mice were administered with PLX3397 for depleting microglia. Primary microglia and astrocytes were used for in vitro experiments. Transmission electron microscopy analysis and immunostaining were performed to verify the positive effects of CeNP in remyelination and OPC differentiation. Flow cytometry, real-time polymerase chain reaction, immunofluorescence and western blotting were used to detect microglia polarization, astrocyte alteration, and the underlying molecular mechanisms. Results CeNP treatment strongly inhibited ROS-induced NF-κB p65 translocation in both microglia and astrocytes, and significantly decreased the expression of M1 microglia and A1 astrocyte. Furthermore, we found that CeNP treatment promoted remyelination and OPC differentiation after ICH, and such effects were alleviated after microglial depletion. Interestingly, we also found that the number of mature oligodendrocytes was moderately increased in ICH + CeNP + PLX3397-treated mice compared to the ICH + vehicle + PLX3397 group. Therefore, astrocytes might participate in the pathophysiological process. The subsequent phagocytosis assay indicated that A1 astrocyte highly expressed C3, which could bind with microglia C3aR and hinder microglial engulfment of myelin debris. This result further replenished the feedback mechanism from astrocytes to microglia. Conclusion The present study reveals a new mechanism in white matter injury after ICH: ICH induces M1 microglia and A1 astrocyte through ROS-induced NF-κB p65 translocation that hinders OPC maturation. Subsequently, A1 astrocytes inhibit microglial phagocytosis of myelin debris via an astrocytic C3-microglial C3aR axis. Polyethylene glycol-CeNP treatment inhibits this pathological process and ultimately promotes remyelination. Such findings enlighten us that astrocytes and microglia should be regarded as a functional unit in future works. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02101-6.
Collapse
|
27
|
Lee JW, Lee IH, Iimura T, Kong SW. Two macrophages, osteoclasts and microglia: from development to pleiotropy. Bone Res 2021; 9:11. [PMID: 33568650 PMCID: PMC7875961 DOI: 10.1038/s41413-020-00134-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Tissue-resident macrophages are highly specialized to their tissue-specific microenvironments, activated by various inflammatory signals and modulated by genetic and environmental factors. Osteoclasts and microglia are distinct tissue-resident cells of the macrophage lineage in bone and brain that are responsible for pathological changes in osteoporosis and Alzheimer’s disease (AD), respectively. Osteoporosis is more frequently observed in individuals with AD compared to the prevalence in general population. Diagnosis of AD is often delayed until underlying pathophysiological changes progress and cause irreversible damages in structure and function of brain. As such earlier diagnosis and intervention of individuals at higher risk would be indispensable to modify clinical courses. Pleiotropy is the phenomenon that a genetic variant affects multiple traits and the genetic correlation between two traits could suggest a shared molecular mechanism. In this review, we discuss that the Pyk2-mediated actin polymerization pathway in osteoclasts and microglia in bone and brain, respectively, is the horizontal pleiotropic mediator of shared risk factors for osteoporosis and AD.
Collapse
Affiliation(s)
- Ji-Won Lee
- Department of Nephrology, Transplant Research Program, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Pharmacology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586, Japan
| | - In-Hee Lee
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Tadahiro Iimura
- Department of Pharmacology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586, Japan
| | - Sek Won Kong
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
28
|
Salmina AB, Gorina YV, Erofeev AI, Balaban PM, Bezprozvanny IB, Vlasova OL. Optogenetic and chemogenetic modulation of astroglial secretory phenotype. Rev Neurosci 2021; 32:459-479. [PMID: 33550788 DOI: 10.1515/revneuro-2020-0119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 11/28/2020] [Indexed: 12/20/2022]
Abstract
Astrocytes play a major role in brain function and alterations in astrocyte function that contribute to the pathogenesis of many brain disorders. The astrocytes are attractive cellular targets for neuroprotection and brain tissue regeneration. Development of novel approaches to monitor and to control astroglial function is of great importance for further progress in basic neurobiology and in clinical neurology, as well as psychiatry. Recently developed advanced optogenetic and chemogenetic techniques enable precise stimulation of astrocytes in vitro and in vivo, which can be achieved by the expression of light-sensitive channels and receptors, or by expression of receptors exclusively activated by designer drugs. Optogenetic stimulation of astrocytes leads to dramatic changes in intracellular calcium concentrations and causes the release of gliotransmitters. Optogenetic and chemogenetic protocols for astrocyte activation aid in extracting novel information regarding the function of brain's neurovascular unit. This review summarizes current data obtained by this approach and discusses a potential mechanistic connection between astrocyte stimulation and changes in brain physiology.
Collapse
Affiliation(s)
- Alla B Salmina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Yana V Gorina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Alexander I Erofeev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Pavel M Balaban
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Ilya B Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Olga L Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
29
|
Lana D, Ugolini F, Giovannini MG. Space-Dependent Glia-Neuron Interplay in the Hippocampus of Transgenic Models of β-Amyloid Deposition. Int J Mol Sci 2020; 21:E9441. [PMID: 33322419 PMCID: PMC7763751 DOI: 10.3390/ijms21249441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
This review is focused on the description and discussion of the alterations of astrocytes and microglia interplay in models of Alzheimer's disease (AD). AD is an age-related neurodegenerative pathology with a slowly progressive and irreversible decline of cognitive functions. One of AD's histopathological hallmarks is the deposition of amyloid beta (Aβ) plaques in the brain. Long regarded as a non-specific, mere consequence of AD pathology, activation of microglia and astrocytes is now considered a key factor in both initiation and progression of the disease, and suppression of astrogliosis exacerbates neuropathology. Reactive astrocytes and microglia overexpress many cytokines, chemokines, and signaling molecules that activate or damage neighboring cells and their mutual interplay can result in virtuous/vicious cycles which differ in different brain regions. Heterogeneity of glia, either between or within a particular brain region, is likely to be relevant in healthy conditions and disease processes. Differential crosstalk between astrocytes and microglia in CA1 and CA3 areas of the hippocampus can be responsible for the differential sensitivity of the two areas to insults. Understanding the spatial differences and roles of glia will allow us to assess how these interactions can influence the state and progression of the disease, and will be critical for identifying therapeutic strategies.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| |
Collapse
|
30
|
Bozic I, Savic D, Lavrnja I. Astrocyte phenotypes: Emphasis on potential markers in neuroinflammation. Histol Histopathol 2020; 36:267-290. [PMID: 33226087 DOI: 10.14670/hh-18-284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes, the most abundant glial cells in the central nervous system (CNS), have numerous integral roles in all CNS functions. They are essential for synaptic transmission and support neurons by providing metabolic substrates, secreting growth factors and regulating extracellular concentrations of ions and neurotransmitters. Astrocytes respond to CNS insults through reactive astrogliosis, in which they go through many functional and molecular changes. In neuroinflammatory conditions reactive astrocytes exert both beneficial and detrimental functions, depending on the context and heterogeneity of astrocytic populations. In this review we profile astrocytic diversity in the context of neuroinflammation; with a specific focus on multiple sclerosis (MS) and its best-described animal model experimental autoimmune encephalomyelitis (EAE). We characterize two main subtypes, protoplasmic and fibrous astrocytes and describe the role of intermediate filaments in the physiology and pathology of these cells. Additionally, we outline a variety of markers that are emerging as important in investigating astrocytic biology in both physiological conditions and neuroinflammation.
Collapse
Affiliation(s)
- Iva Bozic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Savic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
31
|
Lana D, Ugolini F, Giovannini MG. An Overview on the Differential Interplay Among Neurons-Astrocytes-Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front Cell Neurosci 2020; 14:585833. [PMID: 33262692 PMCID: PMC7686560 DOI: 10.3389/fncel.2020.585833] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons have been long regarded as the basic functional cells of the brain, whereas astrocytes and microglia have been regarded only as elements of support. However, proper intercommunication among neurons-astrocytes-microglia is of fundamental importance for the functional organization of the brain. Perturbation in the regulation of brain energy metabolism not only in neurons but also in astrocytes and microglia may be one of the pathophysiological mechanisms of neurodegeneration, especially in hypoxia/ischemia. Glial activation has long been considered detrimental for survival of neurons, but recently it appears that glial responses to an insult are not equal but vary in different brain areas. In this review, we first take into consideration the modifications of the vascular unit of the glymphatic system and glial metabolism in hypoxic conditions. Using the method of triple-labeling fluorescent immunohistochemistry coupled with confocal microscopy (TIC), we recently studied the interplay among neurons, astrocytes, and microglia in chronic brain hypoperfusion. We evaluated the quantitative and morpho-functional alterations of the neuron-astrocyte-microglia triads comparing the hippocampal CA1 area, more vulnerable to ischemia, to the CA3 area, less vulnerable. In these contiguous and interconnected areas, in the same experimental hypoxic conditions, astrocytes and microglia show differential, finely regulated, region-specific reactivities. In both areas, astrocytes and microglia form triad clusters with apoptotic, degenerating neurons. In the neuron-astrocyte-microglia triads, the cell body of a damaged neuron is infiltrated and bisected by branches of astrocyte that create a microscar around it while a microglial cell phagocytoses the damaged neuron. These coordinated actions are consistent with the scavenging and protective activities of microglia. In hypoxia, the neuron-astrocyte-microglia triads are more numerous in CA3 than in CA1, further indicating their protective effects. These data, taken from contiguous and interconnected hippocampal areas, demonstrate that glial response to the same hypoxic insult is not equal but varies significantly. Understanding the differences of glial reactivity is of great interest to explain the differential susceptibility of hippocampal areas to hypoxia/ischemia. Further studies may evidence the differential reactivity of glia in different brain areas, explaining the higher or lower sensitivity of these areas to different insults and whether glia may represent a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
32
|
Blest‐Hopley G, O'Neill A, Wilson R, Giampietro V, Lythgoe D, Egerton A, Bhattacharyya S. Adolescent-onset heavy cannabis use associated with significantly reduced glial but not neuronal markers and glutamate levels in the hippocampus. Addict Biol 2020; 25:e12827. [PMID: 31478302 DOI: 10.1111/adb.12827] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022]
Abstract
Cannabis use has been associated with adverse mental health outcomes, the neurochemical underpinnings of which are poorly understood. Although preclinical evidence suggests glutamatergic dysfunction following cannabis exposure in several brain regions including the hippocampus, evidence from human studies have been inconsistent. We investigated the effect of persistent cannabis use on the brain levels of N-acetyl aspartate (NAA) and myoinositol, the metabolite markers of neurons and glia, the site of the main central cannabinoid CB1 receptor, and the levels of glutamate, the neurotransmitter directly affected by CB1 modulation. We investigated cannabis users (CUs) who started using during adolescence, the period of greatest vulnerability to cannabis effects and focused on the hippocampus, where type 1 cannabinoid receptors (CBR1) are expressed in high density and have been linked to altered glutamatergic neurotransmission. Twenty-two adolescent-onset CUs and 21 nonusing controls (NU), completed proton magnetic resonance spectroscopy, to measure hippocampal metabolite concentrations. Glutamate, NAA, and myoinositol levels were compared between CU and NU using separate analyses of covariance. CU had significantly lower myoinositol but not glutamate or NAA levels in the hippocampus compared with NU. Myoinositol levels in CU positively correlated with glutamate levels, whereas this association was absent in NU. Altered myoinositol levels may be a marker of glia dysfunction and is consistent with experimental preclinical evidence that cannabinoid-induced glial dysfunction may underlie cannabinoid-induced memory impairments. Future studies using appropriate imaging techniques such as positron emission tomography should investigate whether glial dysfunction associated with cannabis use underlies hippocampal dysfunction and memory impairment in CUs.
Collapse
Affiliation(s)
- Grace Blest‐Hopley
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| | - Aisling O'Neill
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| | - Robin Wilson
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| | - Vincent Giampietro
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| | - David Lythgoe
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| | - Sagnik Bhattacharyya
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
- South London and Maudsley NHS Foundation Trust London UK
| |
Collapse
|
33
|
Colizzi M, Weltens N, Lythgoe DJ, Williams SC, Van Oudenhove L, Bhattacharyya S. Differential sensitivity to the acute psychotomimetic effects of delta-9-tetrahydrocannabinol associated with its differential acute effects on glial function and cortisol. Psychol Med 2020; 52:1-8. [PMID: 33107418 DOI: 10.1017/s0033291720003827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cannabis use has been associated with psychosis through exposure to delta-9-tetrahydrocannabinol (Δ9-THC), its key psychoactive ingredient. Although preclinical and human evidence suggests that Δ9-THC acutely modulates glial function and hypothalamic-pituitary-adrenal (HPA) axis activity, whether differential sensitivity to the acute psychotomimetic effects of Δ9-THC is associated with differential effects of Δ9-THC on glial function and HPA-axis response has never been tested. METHODS A double-blind, randomized, placebo-controlled, crossover study investigated whether sensitivity to the psychotomimetic effects of Δ9-THC moderates the acute effects of a single Δ9-THC dose (1.19 mg/2 ml) on myo-inositol levels, a surrogate marker of glia, in the Anterior Cingulate Cortex (ACC), and circadian cortisol levels, the key neuroendocrine marker of the HPA-axis, in a set of 16 healthy participants (seven males) with modest previous cannabis exposure. RESULTS The Δ9-THC-induced change in ACC myo-inositol levels differed significantly between those sensitive to (Δ9-THC minus placebo; M = -0.251, s.d. = 1.242) and those not sensitive (M = 1.615, s.d. = 1.753) to the psychotomimetic effects of the drug (t(14) = 2.459, p = 0.028). Further, the Δ9-THC-induced change in cortisol levels over the study period (baseline minus 2.5 h post-drug injection) differed significantly between those sensitive to (Δ9-THC minus placebo; M = -275.4, s.d. = 207.519) and those not sensitive (M = 74.2, s.d. = 209.281) to the psychotomimetic effects of the drug (t(13) = 3.068, p = 0.009). Specifically, Δ9-THC exposure lowered ACC myo-inositol levels and disrupted the physiological diurnal cortisol decrease only in those subjects developing transient psychosis-like symptoms. CONCLUSIONS The interindividual differences in transient psychosis-like effects of Δ9-THC are the result of its differential impact on glial function and stress response.
Collapse
Affiliation(s)
- Marco Colizzi
- National Institute for Health Research (NIHR) Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- Section of Psychiatry, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Nathalie Weltens
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, University of Leuven, Belgium
| | - David J Lythgoe
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Steve Cr Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Lukas Van Oudenhove
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, University of Leuven, Belgium
| | - Sagnik Bhattacharyya
- National Institute for Health Research (NIHR) Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, and Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| |
Collapse
|
34
|
Trujillo-Estrada L, Gomez-Arboledas A, Forner S, Martini AC, Gutierrez A, Baglietto-Vargas D, LaFerla FM. Astrocytes: From the Physiology to the Disease. Curr Alzheimer Res 2020; 16:675-698. [PMID: 31470787 DOI: 10.2174/1567205016666190830110152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/12/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
Abstract
Astrocytes are key cells for adequate brain formation and regulation of cerebral blood flow as well as for the maintenance of neuronal metabolism, neurotransmitter synthesis and exocytosis, and synaptic transmission. Many of these functions are intrinsically related to neurodegeneration, allowing refocusing on the role of astrocytes in physiological and neurodegenerative states. Indeed, emerging evidence in the field indicates that abnormalities in the astrocytic function are involved in the pathogenesis of multiple neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD) and Amyotrophic Lateral Sclerosis (ALS). In the present review, we highlight the physiological role of astrocytes in the CNS, including their communication with other cells in the brain. Furthermore, we discuss exciting findings and novel experimental approaches that elucidate the role of astrocytes in multiple neurological disorders.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Angela Gomez-Arboledas
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Stefânia Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Alessandra Cadete Martini
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Antonia Gutierrez
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| |
Collapse
|
35
|
Sanz P, Garcia-Gimeno MA. Reactive Glia Inflammatory Signaling Pathways and Epilepsy. Int J Mol Sci 2020; 21:ijms21114096. [PMID: 32521797 PMCID: PMC7312833 DOI: 10.3390/ijms21114096] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation and epilepsy are interconnected. Brain inflammation promotes neuronal hyper-excitability and seizures, and dysregulation in the glia immune-inflammatory function is a common factor that predisposes or contributes to the generation of seizures. At the same time, acute seizures upregulate the production of pro-inflammatory cytokines in microglia and astrocytes, triggering a downstream cascade of inflammatory mediators. Therefore, epileptic seizures and inflammatory mediators form a vicious positive feedback loop, reinforcing each other. In this work, we have reviewed the main glial signaling pathways involved in neuroinflammation, how they are affected in epileptic conditions, and the therapeutic opportunities they offer to prevent these disorders.
Collapse
Affiliation(s)
- Pascual Sanz
- Instituto de Biomedicina de Valencia (CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Jaime Roig 11, 46010 Valencia, Spain
- Correspondence: ; Tel.: +34-963391779; Fax: +34-963690800
| | - Maria Adelaida Garcia-Gimeno
- Department of Biotechnology, Escuela Técnica Superior de Ingeniería Agronómica y del Medio Natural (ETSIAMN), Universitat Politècnica de València, 46022 Valencia, Spain;
| |
Collapse
|
36
|
Kim DY, Zhang H, Park S, Kim Y, Bae CR, Kim YM, Kwon YG. CU06-1004 (endothelial dysfunction blocker) ameliorates astrocyte end-feet swelling by stabilizing endothelial cell junctions in cerebral ischemia/reperfusion injury. J Mol Med (Berl) 2020; 98:875-886. [PMID: 32415357 PMCID: PMC7297708 DOI: 10.1007/s00109-020-01920-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/27/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023]
Abstract
Cerebral ischemia, or stroke, is widespread leading cause of death and disability. Surgical and pharmacological interventions that recover blood flow are the most effective treatment strategies for stroke patients. However, restoring the blood supply is accompanied by severe reperfusion injury, with edema and astrocyte end-feet disruption. Here, we report that the oral administration of CU06-1004 (previously Sac-1004), immediately after onset of ischemia/reperfusion (I/R), ameliorated cerebral damage. CU06-1004 stabilized blood‑brain barrier by inhibiting the disruption of the tight junction-related protein zona occludens-1 and the cortical actin ring in endothelial cells (ECs) after I/R. Interestingly, CU06-1004 significantly suppressed astrocyte end-feet swelling following I/R, by reducing aquaporin 4 and connexin 43 levels, which mediates swelling. Furthermore, the degradation of β1-integrin and β-dystroglycan, which anchors to the cortical actin ring in ECs, was inhibited by CU06-1004 administration after I/R. Consistently, CU06-1004 administration following I/R also suppressed the loss of laminin and collagen type IV, which bind to the cortical actin ring anchoring proteins. Unlike the protective effects of CU06-1004 in ECs, astrocyte viability and proliferation were not directly affected. Taken together, our observations suggest that CU06-1004 inhibits I/R-induced cerebral edema and astrocyte end-feet swelling by maintaining EC junction stability. KEY MESSAGES: • CU06-1004 ameliorates I/R-induced cerebral injury. • EC junction integrity was stabilized by CU06-1004 treatment after I/R. • CU06-1004 reduces astrocyte end-feet swelling following I/R. • EC junction stability affects astrocyte end-feet structure maintenance after I/R.
Collapse
Affiliation(s)
- Dong Young Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Haiying Zhang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- CURACLE Co., Ltd, Gyeonggi-do, Seongnam-si, Republic of Korea
| | - Songyi Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yeaji Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Cho-Rong Bae
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Gangwon-do, Chuncheon-si, Republic of Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
37
|
Ponroy Bally B, Farmer WT, Jones EV, Jessa S, Kacerovsky JB, Mayran A, Peng H, Lefebvre JL, Drouin J, Hayer A, Ernst C, Murai KK. Human iPSC-derived Down syndrome astrocytes display genome-wide perturbations in gene expression, an altered adhesion profile, and increased cellular dynamics. Hum Mol Genet 2020; 29:785-802. [PMID: 31943018 PMCID: PMC7104679 DOI: 10.1093/hmg/ddaa003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/31/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Down syndrome (DS), caused by the triplication of human chromosome 21, leads to significant alterations in brain development and is a major genetic cause of intellectual disability. While much is known about changes to neurons in DS, the effects of trisomy 21 on non-neuronal cells such as astrocytes are poorly understood. Astrocytes are critical for brain development and function, and their alteration may contribute to DS pathophysiology. To better understand the impact of trisomy 21 on astrocytes, we performed RNA-sequencing on astrocytes from newly produced DS human induced pluripotent stem cells (hiPSCs). While chromosome 21 genes were upregulated in DS astrocytes, we found consistent up- and down-regulation of genes across the genome with a strong dysregulation of neurodevelopmental, cell adhesion and extracellular matrix molecules. ATAC (assay for transposase-accessible chromatin)-seq also revealed a global alteration in chromatin state in DS astrocytes, showing modified chromatin accessibility at promoters of cell adhesion and extracellular matrix genes. Along with these transcriptomic and epigenomic changes, DS astrocytes displayed perturbations in cell size and cell spreading as well as modifications to cell-cell and cell-substrate recognition/adhesion, and increases in cellular motility and dynamics. Thus, triplication of chromosome 21 is associated with genome-wide transcriptional, epigenomic and functional alterations in astrocytes that may contribute to altered brain development and function in DS.
Collapse
Affiliation(s)
- Blandine Ponroy Bally
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - W Todd Farmer
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - Emma V Jones
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - Selin Jessa
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
- Quantitative Life Sciences, McGill University, Montreal, QC H3A 2A7, Canada
| | - J Benjamin Kacerovsky
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - Alexandre Mayran
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Huashan Peng
- Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
- Department of Human Genetics, McGill University, Montreal, QC H4H 1R3, Canada
- Douglas Hospital Research Institute, Verdun, QC H4H 1R3, Canada
| | - Julie L Lefebvre
- Department of Molecular Genetics, Program for Neuroscience and Mental Health, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Jacques Drouin
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Arnold Hayer
- Department of Biology, McGill University, Bellini Life Sciences Complex, Montreal, QC H3G 0B1, Canada
| | - Carl Ernst
- Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
- Department of Human Genetics, McGill University, Montreal, QC H4H 1R3, Canada
- Douglas Hospital Research Institute, Verdun, QC H4H 1R3, Canada
| | - Keith K Murai
- Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
- Quantitative Life Sciences, McGill University, Montreal, QC H3A 2A7, Canada
| |
Collapse
|
38
|
Zarei-Kheirabadi M, Vaccaro AR, Rahimi-Movaghar V, Kiani S, Baharvand H. An Overview of Extrinsic and Intrinsic Mechanisms Involved in Astrocyte Development in the Central Nervous System. Stem Cells Dev 2020; 29:266-280. [PMID: 31847709 DOI: 10.1089/scd.2019.0189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the past few decades, our knowledge about the function of the central nervous system (CNS) and astrocytes has improved, and research has confirmed the key roles that astrocytes play in the physiology and pathology of the CNS. Here, we reviewed the intrinsic and extrinsic mechanisms that regulate the development of astrocytes, which are generated from radial glial cells. These regulatory systems modulate various signaling pathways and transcription factors. In this review, four stages of astrocyte development-specification (patterning and switch), migration, proliferation, and maturation, are discussed. In astrocyte patterning, VA1-VA3 domains create the astrocyte subtypes by differential expression of Slit1 and Reelin in the spinal cord. In the brain, patterning creates several astrocyte subtypes by different organizing centers. At the switch step, the janus kinase-signal transducer and activator of transcription pathway governs the transition of neurogenesis to gliogenesis. Bone marrow protein and Notch pathways are also important players of the progliogenic switch. Intrinsic regulation is mediated by DNA methylation transferases, and polycomb group complexes can intrinsically affect the development of astrocytes. In the next stage, these cells proliferate and migrate to their final location. Astrocyte maturation is accomplished through the development of cellular processes, molecular markers, and functions.
Collapse
Affiliation(s)
- Masoumeh Zarei-Kheirabadi
- Department of Brain, Cognitive Sciences and Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Alexander R Vaccaro
- Department of Orthopedics, Rothman Orthopedic Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Kiani
- Department of Brain, Cognitive Sciences and Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
39
|
Zhu J, Tsai NP. Ubiquitination and E3 Ubiquitin Ligases in Rare Neurological Diseases with Comorbid Epilepsy. Neuroscience 2020; 428:90-99. [DOI: 10.1016/j.neuroscience.2019.12.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022]
|
40
|
Wang G, He X, Zhu G, Li D, Shi J, Zhang F. Ellagic acid supports neuron by regulating astroglia Nrf2. Biotechnol Appl Biochem 2019; 66:738-743. [DOI: 10.1002/bab.1791] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/08/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Guo‐Qing Wang
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Xue‐Mei He
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Guo‐Fu Zhu
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Dai‐Di Li
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Jing‐Shan Shi
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Guizhou Zunyi Medical University Zunyi Guizhou People's Republic of China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
- Key Laboratory of Basic Pharmacology of Ministry of Education Zunyi Medical University Zunyi Guizhou People's Republic of China
| |
Collapse
|
41
|
Xian P, Hei Y, Wang R, Wang T, Yang J, Li J, Di Z, Liu Z, Baskys A, Liu W, Wu S, Long Q. Mesenchymal stem cell-derived exosomes as a nanotherapeutic agent for amelioration of inflammation-induced astrocyte alterations in mice. Am J Cancer Res 2019; 9:5956-5975. [PMID: 31534531 PMCID: PMC6735367 DOI: 10.7150/thno.33872] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/24/2019] [Indexed: 01/02/2023] Open
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exo) have robust anti-inflammatory effects in the treatment of neurological diseases such as epilepsy, stroke, or traumatic brain injury. While astrocytes are thought to be mediators of these effects, their precise role remains poorly understood. To address this issue, we investigated the putative therapeutic effects and mechanism of MSC-Exo on inflammation-induced alterations in astrocytes. Methods: Lipopolysaccharide (LPS)-stimulated hippocampal astrocytes in primary culture were treated with MSC-Exo, which were also administered in pilocarpine-induced status epilepticus (SE) mice. Exosomal integration, reactive astrogliosis, inflammatory responses, calcium signaling, and mitochondrial membrane potentials (MMP) were monitored. To experimentally probe the molecular mechanism of MSC-Exo actions on the inflammation-induced astrocytic activation, we inhibited the nuclear factor erythroid-derived 2, like 2 (Nrf2, a key mediator in neuroinflammation and oxidative stress) by sgRNA (in vitro) or ML385 (Nrf2 inhibitor) in vivo. Results: MSC-Exo were incorporated into hippocampal astrocytes as well as attenuated reactive astrogliosis and inflammatory responses in vitro and in vivo. Also, MSC-Exo ameliorated LPS-induced aberrant calcium signaling and mitochondrial dysfunction in culture, and SE-induced learning and memory impairments in mice. Furthermore, the putative therapeutic effects of MSC-Exo on inflammation-induced astrocytic activation (e.g., reduced reactive astrogliosis, NF-κB deactivation) were weakened by Nrf2 inhibition. Conclusions: Our results show that MSC-Exo ameliorate inflammation-induced astrocyte alterations and that the Nrf2-NF-κB signaling pathway is involved in regulating astrocyte activation in mice. These data suggest the promising potential of MSC-Exo as a nanotherapeutic agent for the treatment of neurological diseases with hippocampal astrocyte alterations.
Collapse
|
42
|
Steardo L, de Filippis R, Carbone EA, Segura-Garcia C, Verkhratsky A, De Fazio P. Sleep Disturbance in Bipolar Disorder: Neuroglia and Circadian Rhythms. Front Psychiatry 2019; 10:501. [PMID: 31379620 PMCID: PMC6656854 DOI: 10.3389/fpsyt.2019.00501] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
The worldwide prevalence of sleep disorders is approximately 50%, with an even higher occurrence in a psychiatric population. Bipolar disorder (BD) is a severe mental illness characterized by shifts in mood and activity. The BD syndrome also involves heterogeneous symptomatology, including cognitive dysfunctions and impairments of the autonomic nervous system. Sleep abnormalities are frequently associated with BD and are often a good predictor of a mood swing. Preservation of stable sleep-wake cycles is therefore a key to the maintenance of stability in BD, indicating the crucial role of circadian rhythms in this syndrome. The symptom most widespread in BD is insomnia, followed by excessive daytime sleepiness, nightmares, difficulty falling asleep or maintaining sleep, poor sleep quality, sleep talking, sleep walking, and obstructive sleep apnea. Alterations in the structure or duration of sleep are reported in all phases of BD. Understanding the role of neuroglia in BD and in various aspects of sleep is in nascent state. Contributions of the different types of glial cells to BD and sleep abnormalities are discussed in this paper.
Collapse
Affiliation(s)
- Luca Steardo
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Renato de Filippis
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Elvira Anna Carbone
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Cristina Segura-Garcia
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Achucarro Center for Neuroscience, IKERBASQUE, Bilbao, Spain
| | - Pasquale De Fazio
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| |
Collapse
|
43
|
Ullah MF, Ahmad A, Bhat SH, Abu-Duhier FM, Barreto GE, Ashraf GM. Impact of sex differences and gender specificity on behavioral characteristics and pathophysiology of neurodegenerative disorders. Neurosci Biobehav Rev 2019; 102:95-105. [DOI: 10.1016/j.neubiorev.2019.04.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/24/2019] [Accepted: 04/04/2019] [Indexed: 01/06/2023]
|
44
|
Verkhratsky A. Astroglial Calcium Signaling in Aging and Alzheimer's Disease. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035188. [PMID: 31110130 DOI: 10.1101/cshperspect.a035188] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Astrocytes are the homeostatic and protective cells of the central nervous system (CNS). In neurological diseases, astrocytes undergo complex changes, which are subclassified into (1) reactive astrogliosis, an evolutionary conserved defensive rearrangement of cellular phenotype aimed at neuroprotection; (2) pathological remodeling, when astrocytes acquire new features driving pathology; and (3) astrodegeneration, which is manifested by astroglial atrophy and loss of homeostatic functions. In aging brains as well as in the brains affected by Alzheimer's disease (AD), astrocytes acquire both atrophic and reactive phenotypes in a region- and disease-stage-dependent manner. Prevalence of atrophy overreactivity, observed in certain brain regions and in terminal stages of the disease, arguably facilitates the development of neurological deficits. Astrocytes exhibit ionic excitability mediated by changes in intracellular concentration of ions, most importantly of Ca2+ and Na+, with intracellular ion dynamics triggered by the activity of neural networks. AD astrocytes associated with senile plaques demonstrate Ca2+ hyperactivity in the form of aberrant Ca2+ oscillations and pathological long-range Ca2+ waves. Astroglial Ca2+ signaling originating from Ca2+ release from the endoplasmic reticulum is a key factor in initiating astrogliotic response; deficient Ca2+ signaling toolkits observed in entorhinal and prefrontal cortices of AD model animals may account for vulnerability of these regions to the pathology.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom.,Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Achucarro Center for Neuroscience, Ikerbasque, Basque Foundation for Science, 48011 Bilbao, Spain
| |
Collapse
|
45
|
Pregnolato S, Chakkarapani E, Isles AR, Luyt K. Glutamate Transport and Preterm Brain Injury. Front Physiol 2019; 10:417. [PMID: 31068830 PMCID: PMC6491644 DOI: 10.3389/fphys.2019.00417] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/27/2019] [Indexed: 12/19/2022] Open
Abstract
Preterm birth complications are the leading cause of child death worldwide and a top global health priority. Among the survivors, the risk of life-long disabilities is high, including cerebral palsy and impairment of movement, cognition, and behavior. Understanding the molecular mechanisms of preterm brain injuries is at the core of future healthcare improvements. Glutamate excitotoxicity is a key mechanism in preterm brain injury, whereby the accumulation of extracellular glutamate damages the delicate immature oligodendrocytes and neurons, leading to the typical patterns of injury seen in the periventricular white matter. Glutamate excitotoxicity is thought to be induced by an interaction between environmental triggers of injury in the perinatal period, particularly cerebral hypoxia-ischemia and infection/inflammation, and developmental and genetic vulnerabilities. To avoid extracellular build-up of glutamate, the brain relies on rapid uptake by sodium-dependent glutamate transporters. Astrocytic excitatory amino acid transporter 2 (EAAT2) is responsible for up to 95% of glutamate clearance, and several lines of evidence suggest that it is essential for brain functioning. While in the adult EAAT2 is predominantly expressed by astrocytes, EAAT2 is transiently upregulated in the immature oligodendrocytes and selected neuronal populations during mid-late gestation, at the peak time for preterm brain injury. This developmental upregulation may interact with perinatal hypoxia-ischemia and infection/inflammation and contribute to the selective vulnerability of the immature oligodendrocytes and neurons in the preterm brain. Disruption of EAAT2 may involve not only altered expression but also impaired function with reversal of transport direction. Importantly, elevated EAAT2 levels have been found in the reactive astrocytes and macrophages of human infant post-mortem brains with severe white matter injury (cystic periventricular leukomalacia), potentially suggesting an adaptive mechanism against excitotoxicity. Interestingly, EAAT2 is suppressed in animal models of acute hypoxic-ischemic brain injury at term, pointing to an important and complex role in newborn brain injuries. Enhancement of EAAT2 expression and transport function is gathering attention as a potential therapeutic approach for a variety of adult disorders and awaits exploration in the context of the preterm brain injuries.
Collapse
Affiliation(s)
- Silvia Pregnolato
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elavazhagan Chakkarapani
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Anthony R Isles
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Karen Luyt
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
46
|
Astroglia in Sepsis Associated Encephalopathy. Neurochem Res 2019; 45:83-99. [PMID: 30778837 PMCID: PMC7089215 DOI: 10.1007/s11064-019-02743-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 01/07/2023]
Abstract
Cellular pathophysiology of sepsis associated encephalopathy (SAE) remains poorly characterised. Brain pathology in SAE, which is manifested by impaired perception, consciousness and cognition, results from multifactorial events, including high levels of systemic cytokines, microbial components and endotoxins, which all damage the brain barriers, instigate neuroinflammation and cause homeostatic failure. Astrocytes, being the principal homeostatic cells of the central nervous system contribute to the brain defence against infection. Forming multifunctional anatomical barriers, astroglial cells maintain brain-systemic interfaces and restrict the damage to the nervous tissue. Astrocytes detect, produce and integrate inflammatory signals between immune cells and cells of brain parenchyma, thus regulating brain immune response. In SAE astrocytes are present in both reactive and astrogliopathic states; balance between these states define evolution of pathology and neurological outcomes. In humans pathophysiology of SAE is complicated by frequent presence of comorbidities, as well as age-related remodelling of the brain tissue with senescence of astroglia; these confounding factors further impact upon SAE progression and neurological deficits.
Collapse
|
47
|
Verkhratsky A, Chvátal A. NMDA Receptors in Astrocytes. Neurochem Res 2019; 45:122-133. [DOI: 10.1007/s11064-019-02750-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/22/2019] [Accepted: 02/05/2019] [Indexed: 12/12/2022]
|
48
|
Verkhratsky A, Ho MS, Vardjan N, Zorec R, Parpura V. General Pathophysiology of Astroglia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:149-179. [PMID: 31583588 PMCID: PMC7188602 DOI: 10.1007/978-981-13-9913-8_7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astroglial cells are involved in most if not in all pathologies of the brain. These cells can change the morpho-functional properties in response to pathology or innate changes of these cells can lead to pathologies. Overall pathological changes in astroglia are complex and diverse and often vary with different disease stages. We classify astrogliopathologies into reactive astrogliosis, astrodegeneration with astroglial atrophy and loss of function, and pathological remodelling of astrocytes. Such changes can occur in neurological, neurodevelopmental, metabolic and psychiatric disorders as well as in infection and toxic insults. Mutation in astrocyte-specific genes leads to specific pathologies, such as Alexander disease, which is a leukodystrophy. We discuss changes in astroglia in the pathological context and identify some molecular entities underlying pathology. These entities within astroglia may repent targets for novel therapeutic intervention in the management of brain pathologies.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
| | - Margaret S Ho
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
- Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
- Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
49
|
Mahan VL. Neurointegrity and neurophysiology: astrocyte, glutamate, and carbon monoxide interactions. Med Gas Res 2019; 9:24-45. [PMID: 30950417 PMCID: PMC6463446 DOI: 10.4103/2045-9912.254639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/15/2019] [Indexed: 12/27/2022] Open
Abstract
Astrocyte contributions to brain function and prevention of neuropathologies are as extensive as that of neurons. Astroglial regulation of glutamate, a primary neurotransmitter, is through uptake, release through vesicular and non-vesicular pathways, and catabolism to intermediates. Homeostasis by astrocytes is considered to be of primary importance in determining normal central nervous system health and central nervous system physiology - glutamate is central to dynamic physiologic changes and central nervous system stability. Gasotransmitters may affect diverse glutamate interactions positively or negatively. The effect of carbon monoxide, an intrinsic central nervous system gasotransmitter, in the complex astrocyte homeostasis of glutamate may offer insights to normal brain development, protection, and its use as a neuromodulator and neurotherapeutic. In this article, we will review the effects of carbon monoxide on astrocyte homeostasis of glutamate.
Collapse
Affiliation(s)
- Vicki L. Mahan
- Division of Pediatric Cardiothoracic Surgery in the Department of Surgery, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
50
|
Scuderi C, Noda M, Verkhratsky A. Editorial: Neuroglia Molecular Mechanisms in Psychiatric Disorders. Front Mol Neurosci 2018; 11:407. [PMID: 30429774 PMCID: PMC6220059 DOI: 10.3389/fnmol.2018.00407] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 01/24/2023] Open
Affiliation(s)
- Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Faculty of Health and Medical Sciences, Center for Basic and Translational Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|