1
|
Li T, Kang X, Zhang S, Wang Y, He J, Li H, Shao C, Kang J. Integrating machine learning and multi-omics analysis to reveal nucleotide metabolism-related immune genes and their functional validation in ischemic stroke. Front Immunol 2025; 16:1561544. [PMID: 40207230 PMCID: PMC11979214 DOI: 10.3389/fimmu.2025.1561544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Background Ischemic stroke (IS) is a major global cause of death and disability, linked to nucleotide metabolism imbalances. This study aimed to identify nucleotide metabolism-related genes associated with IS and explore their roles in disease mechanisms for new diagnostic and therapeutic strategies. Methods IS gene expression data were sourced from the GEO database. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were conducted in R, intersecting results with nucleotide metabolism-related genes. Functional enrichment and connectivity map (cMAP) analyses identified key genes and potential therapeutic agents. Core immune-related genes were determined using LASSO regression, SVM-RFE, and Random Forest algorithms. Immune cell infiltration levels and correlations were analyzed via CIBERSORT. Single-cell RNA sequencing (scRNA-seq) data and molecular docking assessed gene expression, localization, and gene-drug binding. In vivo experiments validated core gene expression. Results Thirty-three candidate genes were identified, mainly involved in immune and inflammatory responses. CFL1, HMCES, and GIMAP1 emerged as key immune-related genes, linked to immune cell infiltration and showing high diagnostic potential. cMAP analysis indicated these genes as drug targets. scRNA-seq clarified their expression and localization, and molecular docking confirmed strong drug binding. In vivo experiments validated their significant expression in IS. Conclusion This study underscores the role of nucleotide metabolism in IS, identifying CFL1, HMCES, and GIMAP1 as potential biomarkers and therapeutic targets, providing insights for IS diagnosis and therapy development.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongyan Li
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, Changchun, China
| | - Chen Shao
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, Changchun, China
| | - Jingsong Kang
- Department of Pathophysiology, Key Laboratory of Pathobiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
2
|
Sequeira RC, Godad A. Understanding Glycogen Synthase Kinase-3: A Novel Avenue for Alzheimer's Disease. Mol Neurobiol 2024; 61:4203-4221. [PMID: 38064104 DOI: 10.1007/s12035-023-03839-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/28/2023] [Indexed: 07/11/2024]
Abstract
Alzheimer's Disease (AD) is the most prevalent form of age-related dementia. Even though a century has passed since the discovery of AD, the exact cause of the disease still remains unknown. As a result, this poses a major hindrance in developing effective therapies for treating AD. Glycogen synthase kinase-3 (GSK-3) is one of the kinases that has been investigated recently as a potential therapeutic target for the treatment of AD. It is also known as human tau protein kinase and is a proline-directed serine-threonine kinase. Since dysregulation of this kinase affects all the major characteristic features of the disease, such as tau phosphorylation, amyloid formation, memory, and synaptic function, it is thought to be a major player in the pathogenesis of AD. In this review, we present the most recent information on the role of this kinase in the onset and progression of AD, as well as significant findings that identify GSK-3 as one of the most important targets for AD therapy. We further discuss the potential of treating AD by targeting GSK-3 and give an overview of the ongoing studies aimed at developing GSK-3 inhibitors in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Ronnita C Sequeira
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Gate No.1, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra, 400056, India
| | - Angel Godad
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Gate No.1, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra, 400056, India.
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India.
| |
Collapse
|
3
|
Wikarska A, Roszak K, Roszek K. Mesenchymal Stem Cells and Purinergic Signaling in Autism Spectrum Disorder: Bridging the Gap between Cell-Based Strategies and Neuro-Immune Modulation. Biomedicines 2024; 12:1310. [PMID: 38927517 PMCID: PMC11201695 DOI: 10.3390/biomedicines12061310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/26/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of autism spectrum disorder (ASD) is still increasing, which means that this neurodevelopmental lifelong pathology requires special scientific attention and efforts focused on developing novel therapeutic approaches. It has become increasingly evident that neuroinflammation and dysregulation of neuro-immune cross-talk are specific hallmarks of ASD, offering the possibility to treat these disorders by factors modulating neuro-immunological interactions. Mesenchymal stem cell-based therapy has already been postulated as one of the therapeutic approaches for ASD; however, less is known about the molecular mechanisms of stem cell influence. One of the possibilities, although still underestimated, is the paracrine purinergic activity of MSCs, by which stem cells ameliorate inflammatory reactions. Modulation of adenosine signaling may help restore neurotransmitter balance, reduce neuroinflammation, and improve overall brain function in individuals with ASD. In our review article, we present a novel insight into purinergic signaling, including but not limited to the adenosinergic pathway and its role in neuroinflammation and neuro-immune cross-talk modulation. We anticipate that by achieving a greater understanding of the purinergic signaling contribution to ASD and related disorders, novel therapeutic strategies may be devised for patients with autism in the near future.
Collapse
Affiliation(s)
| | | | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Torun, Poland; (A.W.); (K.R.)
| |
Collapse
|
4
|
Yoshimoto K, Maki K, Adachi T, Kamei KI. Cyclic Stretching Enhances Angiocrine Signals at Liver Bud Stage from Human Pluripotent Stem Cells in Two-Dimensional Culture. Tissue Eng Part A 2024; 30:426-439. [PMID: 38062736 DOI: 10.1089/ten.tea.2023.0148] [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] [Indexed: 01/12/2024] Open
Abstract
Angiocrine signals during the development and growth of organs, including the liver, intestine, lung, and bone, are essential components of intercellular communication. The signals elicited during the liver bud stage are critical for vascularization and enhanced during the intercellular communication between the cells negative for kinase insert domain receptor (KDR) (KDR- cells) and the cells positive for KDR (KDR+ cells), which constitute the liver bud. However, the use of a human pluripotent stem cell (hPSC)-derived system has not facilitated the generation of a perfusable vascularized liver organoid that allows elucidation of liver development and has great potential for liver transplantation. This is largely owing to the lack of fundamental understanding to induce angiocrine signals in KDR- and KDR+ cells during the liver bud stage. We hypothesized that mechanical stimuli of cyclic stretching/pushing by the fetal heart adjacent to the liver bud could be the main contributor to promoting angiocrine signals in KDR- and KDR+ cells during the liver bud stage. In this study, we show that an organ-on-a-chip platform allows the emulation of an in vivo-like mechanical environment for the liver bud stage in vitro and investigate the role of cyclic mechanical stretching (cMS) to angiocrine signals in KDR- and KDR+ cells derived from hPSCs. RNA sequencing revealed that the expression of genes associated with epithelial-to-mesenchymal transition, including angiocrine signals, such as hepatocyte growth factor (HGF) and matrix metallopeptidase 9 (MMP9), were increased by cMS in cocultured KDR- and KDR+ cells. The expression and secretions of HGF and MMP9 were increased by 1.98- and 1.69-fold and 3.23- and 3.72-fold with cMS in the cocultured KDR- and KDR+ cells but were not increased by cMS in the monocultured KDR- and KDR+ cells, respectively. Finally, cMS during the liver bud stage did not lead to the dedifferentiation of hepatocytes, as the cells with cMS showed hepatic maker expression (CYP3A4, CYP3A7, ALB, and AAT) and 1.71-fold higher CYP3A activity than the cells without cMS, during 12 day-hepatocyte maturation after halting cMS. Our findings provide new insights into the mechanical factors during the liver bud stage and directions for future improvements in the engineered liver tissue.
Collapse
Affiliation(s)
- Koki Yoshimoto
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Koichiro Maki
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Taiji Adachi
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Liaoning, China
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Liaoning, China
- Programs of Biology and Bioengineering, Divisions of Science and Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, Brooklyn, New York, USA
| |
Collapse
|
5
|
Markitantova Y, Fokin A, Boguslavsky D, Simirskii V, Kulikov A. Molecular Signatures Integral to Natural Reprogramming in the Pigment Epithelium Cells after Retinal Detachment in Pleurodeles waltl. Int J Mol Sci 2023; 24:16940. [PMID: 38069262 PMCID: PMC10707686 DOI: 10.3390/ijms242316940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
The reprogramming of retinal pigment epithelium (RPE) cells into retinal cells (transdifferentiation) lies in the bases of retinal regeneration in several Urodela. The identification of the key genes involved in this process helps with looking for approaches to the prevention and treatment of RPE-related degenerative diseases of the human retina. The purpose of our study was to examine the transcriptome changes at initial stages of RPE cell reprogramming in adult newt Pleurodeles waltl. RPE was isolated from the eye samples of day 0, 4, and 7 after experimental surgical detachment of the neural retina and was used for a de novo transcriptome assembly through the RNA-Seq method. A total of 1019 transcripts corresponding to the differently expressed genes have been revealed in silico: the 83 increased the expression at an early stage, and 168 increased the expression at a late stage of RPE reprogramming. We have identified up-regulation of classical early response genes, chaperones and co-chaperones, genes involved in the regulation of protein biosynthesis, suppressors of oncogenes, and EMT-related genes. We revealed the growth in the proportion of down-regulated ribosomal and translation-associated genes. Our findings contribute to revealing the molecular mechanism of RPE reprogramming in Urodela.
Collapse
Affiliation(s)
| | | | | | - Vladimir Simirskii
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (Y.M.); (A.K.)
| | | |
Collapse
|
6
|
Tien TY, Wu YJ, Su CH, Hsieh CL, Wang BJ, Lee YN, Su Y, Yeh HI. Pannexin 1 Modulates Angiogenic Activities of Human Endothelial Colony-Forming Cells Through IGF-1 Mechanism and Is a Marker of Senescence. Arterioscler Thromb Vasc Biol 2023; 43:1935-1951. [PMID: 37589139 DOI: 10.1161/atvbaha.123.319529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND We examined the role of Panxs (pannexins) in human endothelial progenitor cell (EPC) senescence. METHODS Young and replication-induced senescent endothelial colony-forming cells (ECFCs) derived from human circulating EPCs were used to examine cellular activities and senescence-associated indicators after transfection of short interference RNA specific to Panx1 or lentivirus-mediated Panx1 overexpression. Hind limb ischemia mice were used as in vivo angiogenesis model. Protein and phospho-kinase arrays were used to determine underlying mechanisms. RESULTS Panx1 was the predominant Panx isoform in human ECFCs and upregulated in both replication-induced senescent ECFCs and circulating EPCs from aged mice and humans. Cellular activities of the young ECFCs were enhanced by Panx1 downregulation but attenuated by its upregulation. In addition, reduction of Panx1 in the senescent ECFCs could rejuvenate cellular activities with reduced senescence-associated indicators, including senescence-associated β-galactosidase activity, p16INK4a (cyclin-dependent kinase inhibitor 2A), p21 (cyclin-dependent kinase inhibitor 1), acetyl-p53 (tumor protein P53), and phospho-histone H2A.X (histone family member X). In mouse ischemic hind limbs injected senescent ECFCs, blood perfusion ratio, salvaged limb outcome, and capillary density were all improved by Panx1 knockdown. IGF-1 (insulin-like growth factor 1) was significantly increased in the supernatant from senescent ECFCs after Panx1 knockdown. The enhanced activities and paracrine effects of Panx1 knockdown senescent ECFCs were completely inhibited by anti-IGF-1 antibodies. FAK (focal adhesion kinase), ERK (extracellular signal-regulated kinase), and STAT3 (signal transducer and activator of transcription 3) were activated in senescent ECFCs with Panx1 knockdown, in which the intracellular calcium level was reduced, and the activation was inhibited by supplemented calcium. The increased IGF-1 in Panx1-knockdown ECFCs was abrogated, respectively, by inhibitors of FAK (PF562271), ERK (U0126), and STAT3 (NSC74859) and supplemented calcium. CONCLUSIONS Panx1 expression is upregulated in human ECFCs/EPCs with replication-induced senescence and during aging. Angiogenic potential of senescent ECFCs is improved by Panx1 reduction through increased IGF-1 production via activation of the FAK-ERK axis following calcium influx reduction. Our findings provide new strategies to evaluate EPC activities and rejuvenate senescent EPCs for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Ting-Yi Tien
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan (T.-Y.T., Y.S.)
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yih-Jer Wu
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| | - Cheng-Huang Su
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| | - Chin-Ling Hsieh
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Bo-Jeng Wang
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Nan Lee
- Departments of Medical Research (T.-Y.T., C.-L.H., B.-J.W., Y.-N.L.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yeu Su
- Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan (T.-Y.T., Y.S.)
| | - Hung-I Yeh
- Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-I.Y.)
| |
Collapse
|
7
|
Rodriguez-Jimenez FJ, Jendelova P, Erceg S. The activation of dormant ependymal cells following spinal cord injury. Stem Cell Res Ther 2023; 14:175. [PMID: 37408068 DOI: 10.1186/s13287-023-03395-4] [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: 01/12/2023] [Accepted: 06/02/2023] [Indexed: 07/07/2023] Open
Abstract
Ependymal cells, a dormant population of ciliated progenitors found within the central canal of the spinal cord, undergo significant alterations after spinal cord injury (SCI). Understanding the molecular events that induce ependymal cell activation after SCI represents the first step toward controlling the response of the endogenous regenerative machinery in damaged tissues. This response involves the activation of specific signaling pathways in the spinal cord that promotes self-renewal, proliferation, and differentiation. We review our current understanding of the signaling pathways and molecular events that mediate the SCI-induced activation of ependymal cells by focusing on the roles of some cell adhesion molecules, cellular membrane receptors, ion channels (and their crosstalk), and transcription factors. An orchestrated response regulating the expression of receptors and ion channels fine-tunes and coordinates the activation of ependymal cells after SCI or cell transplantation. Understanding the major players in the activation of ependymal cells may help us to understand whether these cells represent a critical source of cells contributing to cellular replacement and tissue regeneration after SCI. A more complete understanding of the role and function of individual signaling pathways in endogenous spinal cord progenitors may foster the development of novel targeted therapies to induce the regeneration of the injured spinal cord.
Collapse
Affiliation(s)
- Francisco Javier Rodriguez-Jimenez
- Stem Cell Therapies in Neurodegenerative Diseases Lab, Research Center "Principe Felipe", C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain.
| | - Pavla Jendelova
- Department of Neuroregeneration, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Slaven Erceg
- Stem Cell Therapies in Neurodegenerative Diseases Lab, Research Center "Principe Felipe", C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain.
- National Stem Cell Bank - Valencia Node, Research Center "Principe Felipe", C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain.
- Department of Neuroregeneration, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
8
|
Rachamalla M, Salahinejad A, Khan M, Datusalia AK, Niyogi S. Chronic dietary exposure to arsenic at environmentally relevant concentrations impairs cognitive performance in adult zebrafish (Danio rerio) via oxidative stress and dopaminergic dysfunction. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 886:163771. [PMID: 37164085 DOI: 10.1016/j.scitotenv.2023.163771] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/12/2023]
Abstract
The current study was designed to evaluate the effects of chronic dietary arsenic exposure on the cognitive performance of adult zebrafish and uncover probable pathways by which arsenic mediates such neurotoxic effects. Adult zebrafish were treated with 3 different dietary arsenic concentrations (30, 60, and 100 μg/g dry weight (dw), as arsenite) in addition to control for 60 days. A latent learning paradigm, which employs a complex maze, was used to assess the cognitive performance of fish. Our results demonstrated that dietary treatment with arsenic, especially at medium (60 μg/g dw) and high (100 μg/g dw) exposure dose levels, significantly impaired the performance of fish in various latent learning tasks evaluated in the present study. Concomitant with cognitive dysfunction, chronic dietary exposure to arsenic was also found to increase arsenic accumulation and dopamine levels, and induce oxidative stress (reduced thiol redox, increased lipid peroxidation and expression of antioxidant enzyme genes) in the brain of zebrafish in a dose-dependent manner. Dopaminergic system in the brain is known to play a critical role in regulating cognitive behaviours in fish, and our observations suggested that chronic dietary treatment with medium and high arsenic doses leads to significant alterations in the expression of genes involved in dopamine signalling (dopamine receptors), synthesis (thyroxine hydroxylase) and metabolism (monoamine oxidase) in the zebrafish brain. Moreover, we also recorded significant downregulation of genes such as the brain-derived neurotrophic factor (BDNF) and ectonucleotidases (entpd2_mg, entpd2_mq, and 5'-nucleotidase), which are critical for learning and memory functions, in the zebrafish brain following chronic dietary exposure to arsenic. Overall, the present study suggests that chronic environmentally relevant dietary exposure to arsenic can impair the cognitive performance in zebrafish, essentially by inducing oxidative stress and disrupting the dopaminergic neurotransmission in the brain.
Collapse
Affiliation(s)
- Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada.
| | - Arash Salahinejad
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Maria Khan
- College of Kinesiology, University of Saskatchewan, Saskatoon, SK S7N 5B2, Canada
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, SK S7N 5B3, Canada
| |
Collapse
|
9
|
Ma XY, Yang TT, Liu L, Peng XC, Qian F, Tang FR. Ependyma in Neurodegenerative Diseases, Radiation-Induced Brain Injury and as a Therapeutic Target for Neurotrophic Factors. Biomolecules 2023; 13:754. [PMID: 37238624 PMCID: PMC10216700 DOI: 10.3390/biom13050754] [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: 03/01/2023] [Revised: 04/03/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The neuron loss caused by the progressive damage to the nervous system is proposed to be the main pathogenesis of neurodegenerative diseases. Ependyma is a layer of ciliated ependymal cells that participates in the formation of the brain-cerebrospinal fluid barrier (BCB). It functions to promotes the circulation of cerebrospinal fluid (CSF) and the material exchange between CSF and brain interstitial fluid. Radiation-induced brain injury (RIBI) shows obvious impairments of the blood-brain barrier (BBB). In the neuroinflammatory processes after acute brain injury, a large amount of complement proteins and infiltrated immune cells are circulated in the CSF to resist brain damage and promote substance exchange through the BCB. However, as the protective barrier lining the brain ventricles, the ependyma is extremely vulnerable to cytotoxic and cytolytic immune responses. When the ependyma is damaged, the integrity of BCB is destroyed, and the CSF flow and material exchange is affected, leading to brain microenvironment imbalance, which plays a vital role in the pathogenesis of neurodegenerative diseases. Epidermal growth factor (EGF) and other neurotrophic factors promote the differentiation and maturation of ependymal cells to maintain the integrity of the ependyma and the activity of ependymal cilia, and may have therapeutic potential in restoring the homeostasis of the brain microenvironment after RIBI or during the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xin-Yu Ma
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Ting-Ting Yang
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Lian Liu
- Department of Pharmacology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Xiao-Chun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng Qian
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng-Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
10
|
Paoletti F, Covaceuszach S, Cassetta A, Calabrese AN, Novak U, Konarev P, Grdadolnik J, Lamba D, Golič Grdadolnik S. Distinct conformational changes occur within the intrinsically unstructured pro-domain of pro-Nerve Growth Factor in the presence of ATP and Mg 2. Protein Sci 2023; 32:e4563. [PMID: 36605018 PMCID: PMC9878617 DOI: 10.1002/pro.4563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/24/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Nerve growth factor (NGF), the prototypical neurotrophic factor, is involved in the maintenance and growth of specific neuronal populations, whereas its precursor, proNGF, is involved in neuronal apoptosis. Binding of NGF or proNGF to TrkA, p75NTR , and VP10p receptors triggers complex intracellular signaling pathways that can be modulated by endogenous small-molecule ligands. Here, we show by isothermal titration calorimetry and NMR that ATP binds to the intrinsically disordered pro-peptide of proNGF with a micromolar dissociation constant. We demonstrate that Mg2+ , known to play a physiological role in neurons, modulates the ATP/proNGF interaction. An integrative structural biophysics analysis by small angle X-ray scattering and hydrogen-deuterium exchange mass spectrometry unveils that ATP binding induces a conformational rearrangement of the flexible pro-peptide domain of proNGF. This suggests that ATP may act as an allosteric modulator of the overall proNGF conformation, whose likely distinct biological activity may ultimately affect its physiological homeostasis.
Collapse
Affiliation(s)
- Francesca Paoletti
- Laboratory for Molecular Structural Dynamics, Theory DepartmentNational Institute of ChemistryLjubljanaSlovenia
| | | | - Alberto Cassetta
- Institute of Crystallography—C.N.R.—Trieste OutstationTriesteItaly
| | - Antonio N. Calabrese
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsUK
| | - Urban Novak
- Laboratory for Molecular Structural Dynamics, Theory DepartmentNational Institute of ChemistryLjubljanaSlovenia
| | - Petr Konarev
- A.V. Shubnikov Institute of Crystallography of Federal Scientific Research Centre “Crystallography and Photonics”Russian Academy of SciencesMoscowRussia
| | - Jože Grdadolnik
- Laboratory for Molecular Structural Dynamics, Theory DepartmentNational Institute of ChemistryLjubljanaSlovenia
| | - Doriano Lamba
- Institute of Crystallography—C.N.R.—Trieste OutstationTriesteItaly
- Interuniversity Consortium “Biostructures and Biosystems National Institute”RomeItaly
| | - Simona Golič Grdadolnik
- Laboratory for Molecular Structural Dynamics, Theory DepartmentNational Institute of ChemistryLjubljanaSlovenia
| |
Collapse
|
11
|
Shen K, Wu D, Sun B, Zhu Y, Wang H, Zou W, Ma Y, Lu Z. Ginsenoside Rg1 promotes astrocyte-to-neuron transdifferentiation in rat and its possible mechanism. CNS Neurosci Ther 2022; 29:256-269. [PMID: 36352836 PMCID: PMC9804042 DOI: 10.1111/cns.14000] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/17/2022] [Accepted: 09/30/2022] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Neuronal loss caused by spinal cord injury (SCI) usually contributes to irreversible motor dysfunction. Promoting neuronal regeneration and functional recovery is vital to the repair of SCI. AIMS Astrocytes, activated by SCI with high proliferative capacity and proximity to neuronal lineage, are considered ideal cells for neuronal regeneration. As previous studies identified several small molecules for the induction of astrocyte-to-neuron, we confirmed that ginsenoside Rg1, a neuroprotective herb, could promote the direct transdifferentiation of astrocyte-to-neuron in rat. METHODS AND RESULTS Immunofluorescence staining showed that 26.0 ± 1.5% of the induced cells exhibited less astroglial properties and more neuronal markers with typical neuronal morphologies, reflecting 20.6 ± 0.9% of cholinergic neurons and 22.3 ± 1.9% of dopaminergic neurons. Western blot and qRT-PCR revealed that the induced cells had better antiapoptotic ability and Rg1-promoted neuronal transdifferentiation of reactive astrocytes might take effect through suppressing Notch/Stat3 signal pathway. In vivo, a revised SCI model treated by Rg1 was confirmed with faster functional recovery and less injury lesion cavity. CONCLUSION In summary, our study provided a novel strategy of direct transdifferentiation of endogenous rat reactive astrocytes into neurons with Rg1 and promotion of neuronal regeneration after SCI.
Collapse
Affiliation(s)
- Kelv Shen
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Duanrong Wu
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Baihan Sun
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Yin Zhu
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Hao Wang
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Wenjun Zou
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Yuhang Ma
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Zhengfeng Lu
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
12
|
Contingent intramuscular boosting of P2XR7 axis improves motor function in transgenic ALS mice. Cell Mol Life Sci 2021; 79:7. [PMID: 34936028 PMCID: PMC8695421 DOI: 10.1007/s00018-021-04070-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/06/2022]
Abstract
Amyotrophic lateral sclerosis is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons and severe muscle atrophy without effective treatment. Most research on the disease has been focused on studying motor neurons and supporting cells of the central nervous system. Strikingly, the recent observations have suggested that morpho-functional alterations in skeletal muscle precede motor neuron degeneration, bolstering the interest in studying muscle tissue as a potential target for the delivery of therapies. We previously showed that the systemic administration of the P2XR7 agonist, 2′(3′)-O‐(4-benzoylbenzoyl) adenosine 5-triphosphate (BzATP), enhanced the metabolism and promoted the myogenesis of new fibres in the skeletal muscles of SOD1G93A mice. Here we further corroborated this evidence showing that intramuscular administration of BzATP improved the motor performance of ALS mice by enhancing satellite cells and the muscle pro-regenerative activity of infiltrating macrophages. The preservation of the skeletal muscle retrogradely propagated along with the motor unit, suggesting that backward signalling from the muscle could impinge on motor neuron death. In addition to providing the basis for a suitable adjunct multisystem therapeutic approach in ALS, these data point out that the muscle should be at the centre of ALS research as a target tissue to address novel therapies in combination with those oriented to the CNS.
Collapse
|
13
|
Gupta R, Ambasta RK, Pravir Kumar. Autophagy and apoptosis cascade: which is more prominent in neuronal death? Cell Mol Life Sci 2021; 78:8001-8047. [PMID: 34741624 PMCID: PMC11072037 DOI: 10.1007/s00018-021-04004-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
Autophagy and apoptosis are two crucial self-destructive processes that maintain cellular homeostasis, which are characterized by their morphology and regulated through signal transduction mechanisms. These pathways determine the fate of cellular organelle and protein involved in human health and disease such as neurodegeneration, cancer, and cardiovascular disease. Cell death pathways share common molecular mechanisms, such as mitochondrial dysfunction, oxidative stress, calcium ion concentration, reactive oxygen species, and endoplasmic reticulum stress. Some key signaling molecules such as p53 and VEGF mediated angiogenic pathway exhibit cellular and molecular responses resulting in the triggering of apoptotic and autophagic pathways. Herein, based on previous studies, we describe the intricate relation between cell death pathways through their common genes and the role of various stress-causing agents. Further, extensive research on autophagy and apoptotic machinery excavates the implementation of selective biomarkers, for instance, mTOR, Bcl-2, BH3 family members, caspases, AMPK, PI3K/Akt/GSK3β, and p38/JNK/MAPK, in the pathogenesis and progression of neurodegenerative diseases. This molecular phenomenon will lead to the discovery of possible therapeutic biomolecules as a pharmacological intervention that are involved in the modulation of apoptosis and autophagy pathways. Moreover, we describe the potential role of micro-RNAs, long non-coding RNAs, and biomolecules as therapeutic agents that regulate cell death machinery to treat neurodegenerative diseases. Mounting evidence demonstrated that under stress conditions, such as calcium efflux, endoplasmic reticulum stress, the ubiquitin-proteasome system, and oxidative stress intermediate molecules, namely p53 and VEGF, activate and cause cell death. Further, activation of p53 and VEGF cause alteration in gene expression and dysregulated signaling pathways through the involvement of signaling molecules, namely mTOR, Bcl-2, BH3, AMPK, MAPK, JNK, and PI3K/Akt, and caspases. Alteration in gene expression and signaling cascades cause neurotoxicity and misfolded protein aggregates, which are characteristics features of neurodegenerative diseases. Excessive neurotoxicity and misfolded protein aggregates lead to neuronal cell death by activating death pathways like autophagy and apoptosis. However, autophagy has a dual role in the apoptosis pathways, i.e., activation and inhibition of the apoptosis signaling. Further, micro-RNAs and LncRNAs act as pharmacological regulators of autophagy and apoptosis cascade, whereas, natural compounds and chemical compounds act as pharmacological inhibitors that rescue neuronal cell death through inhibition of apoptosis and autophagic cell death.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
- , Delhi, India.
| |
Collapse
|
14
|
Jahn K, Heese A, Kebir O, Groh A, Bleich S, Krebs MO, Frieling H. Differential Methylation Pattern of Schizophrenia Candidate Genes in Tetrahydrocannabinol-Consuming Treatment-Resistant Schizophrenic Patients Compared to Non-Consumer Patients and Healthy Controls. Neuropsychobiology 2021; 80:36-44. [PMID: 32599581 DOI: 10.1159/000507670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 04/02/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Patients suffering from schizophrenic psychosis show reduced synaptic connectivity compared to healthy individuals. Furthermore, the use of cannabis often precedes the onset of schizophrenic psychosis. Therefore, we investigated whether consumption of cannabis has an impact on the methylation pattern of schizophrenia candidate genes concerned with the development and preservation of synapses and synaptic function. METHODS Fifty blood samples of outpatients affected by treatment-resistant schizophrenic psychosis were collected in the outpatient department of Ch Ste Anne/INSERM (Paris, France). Extracted DNA was sent to the LMN/MHH (Hanover, Germany) where DNA samples were bisulfite converted. The methylation patterns of the promoter region of neuregulin 1 (NRG1), neurexin (NRXN1), disrupted in schizophrenia 1 (DISC1), and microtubule-associated-protein tau (MAPT) were then analysed by sequencing according to Sanger. RESULTS In NRXN1 the group of non-consumer patients showed a methylation rate slightly lower than controls. In patients with preliminary use of tetrahydrocannabinol (THC) the NRXN1 promoter turned out to be methylated almost two times higher than in non-consumer patients. In MAPT, non-consumer patients showed a significant lower mean methylation rate in comparison to controls. In THC-consuming patients the difference compared with controls became less. NRG1 and DISC1 showed no significant differences between groups, whereas DISC1 appeared to be not methylated at all. CONCLUSION In MAPT and NRXN1 mean methylation rates were lower in non-consumer patients compared with controls, which seems to be a compensatory mechanism. With consumption of THC, mean methylation rates were increased: in the case of MAPT compared with controls, and in NRXN1 even significantly beyond that. Methylation of NRG1 and DISC1 seems not to be affected by the psychiatric disorder or by consumption of THC.
Collapse
Affiliation(s)
- Kirsten Jahn
- Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Hannover, Germany,
| | - Astrid Heese
- Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Hannover, Germany
| | - Oussama Kebir
- Centre Hospitalier Sainte Anne (Ch Ste Anne), Paris, France
| | - Adrian Groh
- Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Hannover, Germany
| | - Stefan Bleich
- Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Hannover, Germany
| | | | - Helge Frieling
- Laboratory for Molecular Neurosciences (LMN), Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover (MHH), Hannover, Germany
| |
Collapse
|
15
|
Regnier-Golanov AS, Dündar F, Zumbo P, Betel D, Hernandez MS, Peterson LE, Lo EH, Golanov EV, Britz GW. Hippocampal Transcriptome Changes After Subarachnoid Hemorrhage in Mice. Front Neurol 2021; 12:691631. [PMID: 34354664 PMCID: PMC8329593 DOI: 10.3389/fneur.2021.691631] [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: 04/06/2021] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
After subarachnoid hemorrhage (SAH), up to 95% of surviving patients suffer from post-SAH syndrome, which includes cognitive deficits with impaired memory, executive functions, and emotional disturbances. Although these long-term cognitive deficits are thought to result from damage to temporomesial-hippocampal areas, the underlying mechanisms remain unknown. To fill this gap in knowledge, we performed a systematic RNA sequencing screen of the hippocampus in a mouse model of SAH. SAH was induced by perforation of the circle of Willis in mice. Four days later, hippocampal RNA was obtained from SAH and control (sham perforation) mice. Next-generation RNA sequencing was used to determine differentially expressed genes in the whole bilateral hippocampi remote from the SAH bleeding site. Functional analyses and clustering tools were used to define molecular pathways. Differential gene expression analysis detected 642 upregulated and 398 downregulated genes (false discovery rate <0.10) in SAH compared to Control group. Functional analyses using IPA suite, Gene Ontology terms, REACTOME pathways, and MsigDB Hallmark gene set collections revealed suppression of oligodendrocytes/myelin related genes, and overexpression of genes related to complement system along with genes associated with innate and adaptive immunity, and extracellular matrix reorganization. Interferon regulatory factors, TGF-β1, and BMP were identified as major orchestrating elements in the hippocampal tissue response. The MEME-Suite identified binding motifs of Krüppel-like factors, zinc finger transcription factors, and interferon regulatory factors as overrepresented DNA promoter motifs. This study provides the first systematic gene and pathway database of the hippocampal response after SAH. Our findings suggest that damage of the entorhinal cortex by subarachnoid blood may remotely trigger specific hippocampal responses, which include suppression of oligodendrocyte function. Identification of these novel pathways may allow for development of new therapeutic approaches for post-SAH cognitive deficits.
Collapse
Affiliation(s)
- Angelique S. Regnier-Golanov
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Friederike Dündar
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Paul Zumbo
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States
| | - Magda S. Hernandez
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Leif E. Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX, United States
| | - Eng H. Lo
- Laboratory of Neuroprotection Research, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Eugene V. Golanov
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Gavin W. Britz
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
16
|
P2X7 receptor in multifaceted cellular signalling and its relevance as a potential therapeutic target in different diseases. Eur J Pharmacol 2021; 906:174235. [PMID: 34097884 DOI: 10.1016/j.ejphar.2021.174235] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023]
Abstract
P2X7 receptor, a purinergic receptor family member, is abundantly expressed on many cells, including immune, muscle, bone, neuron, and glia. It acts as an ATP-activated cation channel that permits the influx of Ca2+, Na+ and efflux of K+ ions. The P2X7 receptor plays crucial roles in many physiological processes including cytokine and chemokine secretion, NLRP3 inflammasome activation, cellular growth and differentiation, locomotion, wound healing, transcription factors activation, cell death and T-lymphocyte survival. Past studies have demonstrated the up-regulation and direct association of this receptor in many pathophysiological conditions such as cancer, diabetics, arthritis, tuberculosis (TB) and inflammatory diseases. Hence, targeting this receptor is considered a worthwhile approach to lessen the afflictions associated with the disorders mentioned above by understanding the receptor architecture and downstream signalling processes. Here, in the present review, we have dissected the structural and functional aspects of the P2X7 receptor, emphasizing its role in various diseased conditions. This information will provide in-depth knowledge about the receptor and help to develop apt curative methodologies for the betterment of humanity in the coming years.
Collapse
|
17
|
Chiareli RA, Carvalho GA, Marques BL, Mota LS, Oliveira-Lima OC, Gomes RM, Birbrair A, Gomez RS, Simão F, Klempin F, Leist M, Pinto MCX. The Role of Astrocytes in the Neurorepair Process. Front Cell Dev Biol 2021; 9:665795. [PMID: 34113618 PMCID: PMC8186445 DOI: 10.3389/fcell.2021.665795] [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: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022] Open
Abstract
Astrocytes are highly specialized glial cells responsible for trophic and metabolic support of neurons. They are associated to ionic homeostasis, the regulation of cerebral blood flow and metabolism, the modulation of synaptic activity by capturing and recycle of neurotransmitters and maintenance of the blood-brain barrier. During injuries and infections, astrocytes act in cerebral defense through heterogeneous and progressive changes in their gene expression, morphology, proliferative capacity, and function, which is known as reactive astrocytes. Thus, reactive astrocytes release several signaling molecules that modulates and contributes to the defense against injuries and infection in the central nervous system. Therefore, deciphering the complex signaling pathways of reactive astrocytes after brain damage can contribute to the neuroinflammation control and reveal new molecular targets to stimulate neurorepair process. In this review, we present the current knowledge about the role of astrocytes in brain damage and repair, highlighting the cellular and molecular bases involved in synaptogenesis and neurogenesis. In addition, we present new approaches to modulate the astrocytic activity and potentiates the neurorepair process after brain damage.
Collapse
Affiliation(s)
| | | | | | - Lennia Soares Mota
- Department of Pharmacology, Federal University of Goias, Goiânia, Brazil
| | | | | | - Alexander Birbrair
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Renato Santiago Gomez
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Simão
- Research Division, Vascular Cell Biology, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | | | - Marcel Leist
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | | |
Collapse
|
18
|
Paoletti F, Merzel F, Cassetta A, Ogris I, Covaceuszach S, Grdadolnik J, Lamba D, Golič Grdadolnik S. Endogenous modulators of neurotrophin signaling: Landscape of the transient ATP-NGF interactions. Comput Struct Biotechnol J 2021; 19:2938-2949. [PMID: 34136093 PMCID: PMC8164016 DOI: 10.1016/j.csbj.2021.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 12/02/2022] Open
Abstract
High-resolution solution NMR structure of rhNGF has been determined. Quinary interactions characterize ATP binding to rhNGF. SPR, ITC and STD-NMR reveal ATP binding to rhNGF with mM affinity. NMR and MD analysis pinpoint to the presence of two binding sites of ATP on rhNGF. Stoichiometry of ATP-Mg2+ or Zn2+-rhNGF mixtures affects KD affinity to TrkA/p75NTR.
The Nerve Growth Factor (NGF) neurotrophin acts in the maintenance and growth of neuronal populations. Despite the detailed knowledge of NGF’s role in neuron physiology, the structural and mechanistic determinants of NGF bioactivity modulated by essential endogenous ligands are still lacking. We present the results of an integrated structural and advanced computational approach to characterize the extracellular ATP-NGF interaction. We mapped by NMR the interacting surface and ATP orientation on NGF and revealed the functional role of this interaction in the binding to TrkA and p75NTR receptors by SPR. The role of divalent ions was explored in conjunction with ATP. Our results pinpoint ATP as a likely transient molecular modulator of NGF signaling, in health and disease states.
Collapse
Key Words
- ARIA, Ambiguous Restraints for Iterative Assignment
- ATP modulation
- BDNF, Brain Derived Neurotrophic Factor
- CARA, Computer Aided Resonance Assignment
- CS-E, Chrondroitin Sulfate E
- CSP, Chemical Shift Perturbation
- DSF, Differential Scanning Fluorimetry
- EI-MS, Electron Ionization Mass Spectrometry
- Endogenous ligands
- FGF2, Fibroblast Growth Factor 2
- FT-IR, Fourier Transform Infrared Spectroscopy
- HBD, Heparin Binding Domain
- HSQC, Heteronuclear Single Quantum Coherence
- ITC, Isothermal Titration Calorimetry
- MALDI-TOF MS, Matrix Assisted Laser Desorption Ionization-Time Of Flight Mass Spectrometry
- MD, Molecular Dynamics
- MS, Mass Spectrometry
- NGF interactions
- NGF, Nerve Growth Factor
- NMR, Nuclear Magnetic Resonance
- NOE, Nuclear Overhouser Effect
- NOESY, Nuclear Overhauser Effect Spectroscopy
- NT, NeuroTrophin
- Neurotrophins
- P20, Polysorbate 20
- PME, Particle Mesh Ewald
- RMSD, Root Mean Square Deviation
- SAR, Structure-Activity Relationship
- SPR, Surface Plasmon Resonance
- STD, Saturation-Transfer Difference
- TrkA, Tyrosine Kinase Receptor A
- TrkA, p75NTR receptors
- p75NTR, p75 NeuroTrophin Receptor
- proNGF, proNGF – NGF precursor
- rh-proNGF, recombinant human proNGF – NGF precursor
- rhNGF, recombinant human NGF
- rmNGF, recombinant mouse NGF
Collapse
Affiliation(s)
- Francesca Paoletti
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Franci Merzel
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Alberto Cassetta
- Institute of Crystallography - C.N.R.- Trieste Outstation. Area Science Park - Basovizza, S.S.14 - Km. 163.5, I-34149 Trieste, Italy
| | - Iza Ogris
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Sonia Covaceuszach
- Institute of Crystallography - C.N.R.- Trieste Outstation. Area Science Park - Basovizza, S.S.14 - Km. 163.5, I-34149 Trieste, Italy
| | - Jože Grdadolnik
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Doriano Lamba
- Institute of Crystallography - C.N.R.- Trieste Outstation. Area Science Park - Basovizza, S.S.14 - Km. 163.5, I-34149 Trieste, Italy.,Interuniversity Consortium "Biostructures and Biosystems National Institute", Viale delle Medaglie d'Oro 305, I-00136 Roma, Italy
| | - Simona Golič Grdadolnik
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| |
Collapse
|
19
|
Almeida RF, Ferreira TP, David CVC, Abreu E Silva PC, Dos Santos SA, Rodrigues ALS, Elisabetsky E. Guanine-Based Purines as an Innovative Target to Treat Major Depressive Disorder. Front Pharmacol 2021; 12:652130. [PMID: 33927625 PMCID: PMC8076783 DOI: 10.3389/fphar.2021.652130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/01/2021] [Indexed: 01/18/2023] Open
Affiliation(s)
- Roberto F Almeida
- Departamento de Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, Brazil.,Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tiago P Ferreira
- Departamento de Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Camila V C David
- Departamento de Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Paulo C Abreu E Silva
- Departamento de Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Sulamita A Dos Santos
- Departamento de Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Ana L S Rodrigues
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Elaine Elisabetsky
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
20
|
Synthetic 1,4-Naphthoquinones inhibit P2X7 receptors in murine neuroblastoma cells. Bioorg Med Chem 2021; 31:115975. [PMID: 33401207 DOI: 10.1016/j.bmc.2020.115975] [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: 09/16/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
The P2X7 receptor (P2X7R) is an ATP-gated ion channel and potential therapeutic target for new drug development. In this study, we synthesized a series of new 1,4-naphthoquinone (1,4-NQ) derivatives and investigated their antagonistic effects against mouse P2X7R. We explored the ability of the tested substances to block ATP-induced Ca2+ influx into mouse Neuro-2a cells and selected the four most effective substances: the 1,4-naphthoquinone thioglucosides U-548 and U-557 and their tetracyclic conjugates U-286 and U-556. Biological analysis of these compounds revealed significant in vitro inhibition of murine P2X7R. This inhibition resulted in marked blockade of ethidium bromide (EtBr) and YO-PRO-1 fluorescent dye uptake, pronounced decreases in ROS and NO production and protection of neuronal cell viability against the toxic action of high ATP concentrations. In silico analysis indicated favorable molecular docking results of these 1,4-NQs, pointing to their potential to bind in an allosteric site located in the extracellular region of P2X7R. These findings suggest compounds U-286, U-548, U-556 and U-557 as potential scaffolds for the design of new P2X7R blockers and drugs effective against neuropathic pain and neurodegenerative diseases.
Collapse
|
21
|
Salient brain entities labelled in P2rx7-EGFP reporter mouse embryos include the septum, roof plate glial specializations and circumventricular ependymal organs. Brain Struct Funct 2021; 226:715-741. [PMID: 33427974 PMCID: PMC7981336 DOI: 10.1007/s00429-020-02204-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 12/16/2020] [Indexed: 02/08/2023]
Abstract
The purinergic system is one of the oldest cell-to-cell communication mechanisms and exhibits relevant functions in the regulation of the central nervous system (CNS) development. Amongst the components of the purinergic system, the ionotropic P2X7 receptor (P2X7R) stands out as a potential regulator of brain pathology and physiology. Thus, P2X7R is known to regulate crucial aspects of neuronal cell biology, including axonal elongation, path-finding, synapse formation and neuroprotection. Moreover, P2X7R modulates neuroinflammation and is posed as a therapeutic target in inflammatory, oncogenic and degenerative disorders. However, the lack of reliable technical and pharmacological approaches to detect this receptor represents a major hurdle in its study. Here, we took advantage of the P2rx7-EGFP reporter mouse, which expresses enhanced green fluorescence protein (EGFP) immediately downstream of the P2rx7 proximal promoter, to conduct a detailed study of its distribution. We performed a comprehensive analysis of the pattern of P2X7R expression in the brain of E18.5 mouse embryos revealing interesting areas within the CNS. Particularly, strong labelling was found in the septum, as well as along the entire neural roof plate zone of the brain, except chorioidal roof areas, but including specialized circumventricular roof formations, such as the subfornical and subcommissural organs (SFO; SCO). Moreover, our results reveal what seems a novel circumventricular organ, named by us postarcuate organ (PArcO). Furthermore, this study sheds light on the ongoing debate regarding the specific presence of P2X7R in neurons and may be of interest for the elucidation of additional roles of P2X7R in the idiosyncratic histologic development of the CNS and related systemic functions.
Collapse
|
22
|
Illes P, Xu GY, Tang Y. Purinergic Signaling in the Central Nervous System in Health and Disease. Neurosci Bull 2020; 36:1239-1241. [PMID: 33146814 DOI: 10.1007/s12264-020-00602-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Peter Illes
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.,Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universitaet Leipzig, Leipzig, 04107, Germany
| | - Guang-Yin Xu
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China. .,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
23
|
Calzaferri F, Ruiz-Ruiz C, de Diego AMG, de Pascual R, Méndez-López I, Cano-Abad MF, Maneu V, de Los Ríos C, Gandía L, García AG. The purinergic P2X7 receptor as a potential drug target to combat neuroinflammation in neurodegenerative diseases. Med Res Rev 2020; 40:2427-2465. [PMID: 32677086 DOI: 10.1002/med.21710] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 06/22/2020] [Accepted: 06/27/2020] [Indexed: 12/25/2022]
Abstract
Neurodegenerative diseases (NDDs) represent a huge social burden, particularly in Alzheimer's disease (AD) in which all proposed treatments investigated in murine models have failed during clinical trials (CTs). Thus, novel therapeutic strategies remain crucial. Neuroinflammation is a common pathogenic feature of NDDs. As purinergic P2X7 receptors (P2X7Rs) are gatekeepers of inflammation, they could be developed as drug targets for NDDs. Herein, we review this challenging hypothesis and comment on the numerous studies that have investigated P2X7Rs, emphasizing their molecular structure and functions, as well as their role in inflammation. Then, we elaborate on research undertaken in the field of medicinal chemistry to determine potential P2X7R antagonists. Subsequently, we review the state of neuroinflammation and P2X7R expression in the brain, in animal models and patients suffering from AD, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, multiple sclerosis, and retinal degeneration. Next, we summarize the in vivo studies testing the hypothesis that by mitigating neuroinflammation, P2X7R blockers afford neuroprotection, increasing neuroplasticity and neuronal repair in animal models of NDDs. Finally, we reviewed previous and ongoing CTs investigating compounds directed toward targets associated with NDDs; we propose that CTs with P2X7R antagonists should be initiated. Despite the high expectations for putative P2X7Rs antagonists in various central nervous system diseases, the field is moving forward at a relatively slow pace, presumably due to the complexity of P2X7Rs. A better pharmacological approach to combat NDDs would be a dual strategy, combining P2X7R antagonism with drugs targeting a selective pathway in a given NDD.
Collapse
Affiliation(s)
- Francesco Calzaferri
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Ruiz-Ruiz
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio M G de Diego
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ricardo de Pascual
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Iago Méndez-López
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - María F Cano-Abad
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Victoria Maneu
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, San Vicente del Raspeig, Spain
| | - Cristóbal de Los Ríos
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Gandía
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio G García
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
24
|
Lv G, Li C, Wang W, Li N, Wang K. Silencing SP1 Alleviated Sevoflurane-Induced POCD Development via Cholinergic Anti-inflammatory Pathway. Neurochem Res 2020; 45:2082-2090. [PMID: 32594292 DOI: 10.1007/s11064-020-03070-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 06/03/2020] [Accepted: 06/12/2020] [Indexed: 12/26/2022]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication induced by anesthesia or surgery, which affects the concentration, cognition and memory of patients. Sevoflurane, a clinical anesthetic, could stimulate neuro-inflammation and lead to POCD. Recent studies found that specificity protein 1 (SP1) participates in the development of neurological diseases. Our study aims to elucidate the role of SP1 in sevoflurane-induced POCD pathogenesis. We anesthetized Sprague-Dawley rats and treated the primary hippocampal neurons with sevoflurane to construct the in vivo and in vitro POCD models. Besides, the expression and regulatory mechanism of SP1 in the pathogenesis of POCD were explored. According to the results, sevoflurane anesthesia impaired the cognitive functions of rat, significantly elevated SP1 expression and inactivated the cholinergic anti-inflammatory pathway (CAP) both in vivo and in vitro. Moreover, the sevoflurane-treated rats and neurons also exhibited obvious inflammatory responses and enhanced apoptosis. Loss-of-function assay indicated that SP1 knockdown rescued the deactivation of CAP and alleviated the sevoflurane-induced neuro-inflammation and apoptosis in rat hippocampus. Generally, our study documented that the sevoflurane-induced SP1 up-regulation affected the activation of CAP, leading to the aggravated neuro-inflammation and apoptosis. This may provide a novel sight for POCD therapy.
Collapse
Affiliation(s)
- Guoying Lv
- Department of the First Anesthesiology, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Chuangang Li
- Department of the First Anesthesiology, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Weiwei Wang
- Department of the First Anesthesiology, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Ning Li
- Department of the First Anesthesiology, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Kai Wang
- Department of Hepatology, QiLu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
25
|
Caveolin-1 Regulates P2Y 2 Receptor Signaling during Mechanical Injury in Human 1321N1 Astrocytoma. Biomolecules 2019; 9:biom9100622. [PMID: 31635212 PMCID: PMC6843573 DOI: 10.3390/biom9100622] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022] Open
Abstract
Caveolae-associated protein caveolin-1 (Cav-1) plays key roles in cellular processes such as mechanosensing, receptor coupling to signaling pathways, cell growth, apoptosis, and cancer. In 1321N1 astrocytoma cells Cav-1 interacts with the P2Y2 receptor (P2Y2R) to modulate its downstream signaling. P2Y2R and its signaling machinery also mediate pro-survival actions after mechanical injury. This study determines if Cav-1 knockdown (KD) affects P2Y2R signaling and its pro-survival actions in the 1321N1 astrocytoma cells mechanical injury model system. KD of Cav-1 decreased its expression in 1321N1 cells devoid of or expressing hHAP2Y2R by ~88% and ~85%, respectively. Cav-1 KD had no significant impact on P2Y2R expression. Post-injury densitometric analysis of pERK1/2 and Akt activities in Cav-1-positive 1321N1 cells (devoid of or expressing a hHAP2Y2R) revealed a P2Y2R-dependent temporal increase in both kinases. These temporal increases in pERK1/2 and pAkt were significantly decreased in Cav-1 KD 1321N1 (devoid of or expressing a hHAP2Y2R). Cav-1 KD led to an ~2.0-fold and ~2.4-fold decrease in the magnitude of the hHAP2Y2R-mediated pERK1/2 and pAkt kinases’ activity, respectively. These early-onset hHAP2Y2R-mediated signaling responses in Cav-1-expressing and Cav-1 KD 1321N1 correlated with changes in cell viability (via a resazurin-based method) and apoptosis (via caspase-9 expression). In Cav-1-positive 1321N1 cells, expression of hHAP2Y2R led to a significant increase in cell viability and decreased apoptotic (caspase-9) activity after mechanical injury. In contrast, hHAP2Y2R-elicited changes in viability and apoptotic (caspase-9) activity were decreased after mechanical injury in Cav-1 KD 1321N1 cells expressing hHAP2Y2R. These findings support the importance of Cav-1 in modulating P2Y2R signaling during mechanical injury and its protective actions in a human astrocytoma cell line, whilst shedding light on potential new venues for brain injury or trauma interventions.
Collapse
|
26
|
Sp1 in Astrocyte Is Important for Neurite Outgrowth and Synaptogenesis. Mol Neurobiol 2019; 57:261-277. [PMID: 31317491 DOI: 10.1007/s12035-019-01694-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 07/03/2019] [Indexed: 02/04/2023]
Abstract
In this study, we found that Sp1 was highly expressed in astrocytes, implying that Sp1 might be important for the function of astrocytes. Sp1/GFAP-Cre-ERT2 conditional knockout mice were constructed to study the role of Sp1 in astrocytes. Knockout of Sp1 in astrocytes altered astrocytic morphology and decreased GFAP expression in the cortex and hippocampus but did not affect cell viability. Loss of Sp1 in astrocytes decreased the number of neurons in the cortex and hippocampus. Conditioned medium from primary astrocytes with Sp1 knockout disrupted neuronal dendritic outgrowth and synapse formation, resulting in abnormal learning, memory, and motor behavior. Sp1 knockout in astrocytes altered gene expression, including decreasing the expression of Toll-like receptor 2 and Cfb and increasing the expression of C1q and C4Bp, thereby affecting neurite outgrowth and synapse formation, resulting in disordered neuron function. Studying these gene regulations might be beneficial to understanding neuronal development and brain injury prevention.
Collapse
|
27
|
Reigada D, Calderón-García AÁ, Soto-Catalán M, Nieto-Díaz M, Muñoz-Galdeano T, Del Águila Á, Maza RM. MicroRNA-135a-5p reduces P2X 7 -dependent rise in intracellular calcium and protects against excitotoxicity. J Neurochem 2019; 151:116-130. [PMID: 30924927 DOI: 10.1111/jnc.14700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 01/01/2023]
Abstract
Excitotoxic cell death because of the massive release of glutamate and ATP contributes to the secondary extension of cellular and tissue loss following traumatic spinal cord injury (SCI). Evidence from blockage experiments suggests that over-expression and activation of purinergic receptors, especially P2X7 , produces excitotoxicity in neurodegenerative diseases and trauma of the central nervous system. We hypothesize that the down-regulation of specific miRNAs after the SCI contributes to the over-expression of P2X7 and that restorative strategies can be used to reduce the excitotoxic response. In the present study, we have employed bioinformatic analyses to identify microRNAs whose down-regulation following SCI can be responsible for P2X7 over-expression and excitotoxic activity. Additional luciferase assays validated microRNA-135a-5p (miR-135a) as a posttranscriptional modulator of P2X7 . Moreover, gene expression analysis in spinal cord samples from a rat SCI model confirmed that the decrease in miR-135a expression correlated with P2X7 over-expression after injury. Transfection of cultures of Neuro-2a neuronal cell line with a miR-135a inhibitory sequences (antagomiR-135a), simulating the reduction of miR-135a observed after SCI, resulted in the increase of P2X7 expression and the subsequent ATP-dependent rise in intracellular calcium concentration. Conversely, a restorative strategy employing miR-135a mimicked reduced P2X7 expression, attenuating the increase in intracellular calcium concentration that depends on this receptor and protecting cells from excitotoxic death. Therefore, we conclude that miR-135a is a potential therapeutic target for SCI and that restoration of its expression may reduce the deleterious effects of ATP-dependent excitotoxicity induced after a traumatic spinal cord injury.
Collapse
Affiliation(s)
- David Reigada
- Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla La Mancha (SESCAM), Toledo, Spain
| | - Andrés Ángel Calderón-García
- Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla La Mancha (SESCAM), Toledo, Spain.,Instituto de Neurociencias de Castilla y León (INCYL), Faculty of Medicine, University of Salamanca. Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Manuel Soto-Catalán
- Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla La Mancha (SESCAM), Toledo, Spain
| | - Manuel Nieto-Díaz
- Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla La Mancha (SESCAM), Toledo, Spain
| | - Teresa Muñoz-Galdeano
- Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla La Mancha (SESCAM), Toledo, Spain
| | - Ángela Del Águila
- Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla La Mancha (SESCAM), Toledo, Spain.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rodrigo M Maza
- Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla La Mancha (SESCAM), Toledo, Spain
| |
Collapse
|
28
|
Pérez-Sen R, Queipo MJ, Gil-Redondo JC, Ortega F, Gómez-Villafuertes R, Miras-Portugal MT, Delicado EG. Dual-Specificity Phosphatase Regulation in Neurons and Glial Cells. Int J Mol Sci 2019; 20:ijms20081999. [PMID: 31018603 PMCID: PMC6514851 DOI: 10.3390/ijms20081999] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/19/2019] [Indexed: 01/03/2023] Open
Abstract
Dual-specificity protein phosphatases comprise a protein phosphatase subfamily with selectivity towards mitogen-activated protein (MAP) kinases, also named MKPs, or mitogen-activated protein kinase (MAPK) phosphatases. As powerful regulators of the intensity and duration of MAPK signaling, a relevant role is envisioned for dual-specificity protein phosphatases (DUSPs) in the regulation of biological processes in the nervous system, such as differentiation, synaptic plasticity, and survival. Important neural mediators include nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) that contribute to DUSP transcriptional induction and post-translational mechanisms of DUSP protein stabilization to maintain neuronal survival and differentiation. Potent DUSP gene inducers also include cannabinoids, which preserve DUSP activity in inflammatory conditions. Additionally, nucleotides activating P2X7 and P2Y13 nucleotide receptors behave as novel players in the regulation of DUSP function. They increase cell survival in stressful conditions, regulating DUSP protein turnover and inducing DUSP gene expression. In general terms, in the context of neural cells exposed to damaging conditions, the recovery of DUSP activity is neuroprotective and counteracts pro-apoptotic over-activation of p38 and JNK. In addition, remarkable changes in DUSP function take place during the onset of neuropathologies. The restoration of proper DUSP levels and recovery of MAPK homeostasis underlie the therapeutic effect, indicating that DUSPs can be relevant targets for brain diseases.
Collapse
Affiliation(s)
- Raquel Pérez-Sen
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - María José Queipo
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Juan Carlos Gil-Redondo
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Felipe Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Rosa Gómez-Villafuertes
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - María Teresa Miras-Portugal
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Esmerilda G Delicado
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| |
Collapse
|
29
|
Illes P, Rubini P, Huang L, Tang Y. The P2X7 receptor: a new therapeutic target in Alzheimer’s disease. Expert Opin Ther Targets 2019; 23:165-176. [DOI: 10.1080/14728222.2019.1575811] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Patrizia Rubini
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Lumei Huang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| |
Collapse
|
30
|
Du XJ, Chen YX, Zheng ZC, Wang N, Wang XY, Kong FE. Neural stem cell transplantation inhibits glial cell proliferation and P2X receptor-mediated neuropathic pain in spinal cord injury rats. Neural Regen Res 2019; 14:876-885. [PMID: 30688274 PMCID: PMC6375052 DOI: 10.4103/1673-5374.249236] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
P2X4 and P2X7 receptors play an important role in neuropathic pain after spinal cord injury. Regulation of P2X4 and P2X7 receptors can obviously reduce pain hypersensitivity after injury. To investigate the role of neural stem cell transplantation on P2X receptor-mediated neuropathic pain and explore related mechanisms, a rat model of spinal cord injury was prepared using the free-falling heavy body method with spinal cord segment 10 as the center. Neural stem cells were injected into the injured spinal cord segment using a micro-syringe. Expression levels of P2X4 and P2X7 receptors, neurofilament protein, and glial fibrillary acidic protein were determined by immunohistochemistry and western blot assay. In addition, sensory function was quantitatively assessed by current perception threshold. The Basso-Beattie-Bresnahan locomotor rating scale was used to assess neuropathological pain. The results showed that 4 weeks after neural stem cell transplantation, expression of neurofilament protein in the injured segment was markedly increased, while expression of glial fibrillary acidic protein and P2X4 and P2X7 receptors was decreased. At this time point, motor and sensory functions of rats were obviously improved, and neuropathic pain was alleviated. These findings demonstrated that neural stem cell transplantation reduced overexpression of P2X4 and P2X7 receptors, activated locomotor and sensory function reconstruction, and played an important role in neuropathic pain regulation after spinal cord injury. Therefore, neural stem cell transplantation is one potential option for relieving neuropathic pain mediated by P2X receptors.
Collapse
Affiliation(s)
- Xiao-Jing Du
- Department of Rehabilitation Medicine, the Central Hospital of Taian, Taian, Shandong Province, China
| | - Yue-Xia Chen
- Department of Rehabilitation Medicine, the Central Hospital of Taian, Taian, Shandong Province, China
| | - Zun-Cheng Zheng
- Department of Rehabilitation Medicine, the Central Hospital of Taian, Taian, Shandong Province, China
| | - Nan Wang
- Graduate School, Taishan Medical University, Taian, Shandong Province, China
| | - Xiao-Yu Wang
- Department of Rehabilitation Medicine, the Central Hospital of Taian, Taian, Shandong Province, China
| | - Fan-E Kong
- Graduate School, Taishan Medical University, Taian, Shandong Province, China
| |
Collapse
|
31
|
Miras-Portugal MT, Queipo MJ, Gil-Redondo JC, Ortega F, Gómez-Villafuertes R, Gualix J, Delicado EG, Pérez-Sen R. P2 receptor interaction and signalling cascades in neuroprotection. Brain Res Bull 2018; 151:74-83. [PMID: 30593879 DOI: 10.1016/j.brainresbull.2018.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/27/2018] [Accepted: 12/19/2018] [Indexed: 02/08/2023]
Abstract
Nucleotides can contribute to the survival of different glial and neuronal models at the nervous system via activation of purinergic P2X and P2Y receptors. Their activation counteracts different proapoptotic events, such as excitotoxicity, mitochondrial impairment, oxidative stress and DNA damage, which concur to elicit cell loss in different processes of neurodegeneration and brain injury. Thus, it is frequent to find that different neuroprotective mediators converge in the activation of the same intracellular survival pathways to protect cells from death. The present review focuses on the role of P2Y1 and P2Y13 metabotropic receptors, and P2X7 ionotropic receptors to regulate the balance between survival and apoptosis. In particular, we analyze the intracellular pathways involved in the signaling of these nucleotide receptors to elicit survival, including calcium/PLC, PI3K/Akt/GSK3, MAPK cascades, and the expression of antioxidant and antiapoptotic genes. This review emphasizes the novel contribution of nucleotide receptors to maintain cell homeostasis through the regulation of MAP kinases and phosphatases. Unraveling the different roles found for nucleotide receptors in different models and cellular contexts may be crucial to delineate future therapeutic applications based on targeting nucleotide receptors for neuroprotection.
Collapse
Affiliation(s)
- Mª Teresa Miras-Portugal
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Mª José Queipo
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Juan Carlos Gil-Redondo
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Felipe Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Rosa Gómez-Villafuertes
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Javier Gualix
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Esmerilda G Delicado
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Raquel Pérez-Sen
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| |
Collapse
|
32
|
Duda P, Wiśniewski J, Wójtowicz T, Wójcicka O, Jaśkiewicz M, Drulis-Fajdasz D, Rakus D, McCubrey JA, Gizak A. Targeting GSK3 signaling as a potential therapy of neurodegenerative diseases and aging. Expert Opin Ther Targets 2018; 22:833-848. [PMID: 30244615 DOI: 10.1080/14728222.2018.1526925] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Glycogen synthase kinase 3 (GSK3) is at the center of cellular signaling and controls various aspects of brain functions, including development of the nervous system, neuronal plasticity and onset of neurodegenerative disorders. Areas covered: In this review, recent efforts in elucidating the roles of GSK3 in neuronal plasticity and development of brain pathologies; Alzheimer's and Parkinson's disease, schizophrenia, and age-related neurodegeneration are described. The effect of microglia and astrocytes on development of the pathological states is also discussed. Expert opinion: GSK3β and its signaling pathway partners hold great promise as therapeutic target(s) for a multitude of neurological disorders. Activity of the kinase is often elevated in brain disorders. However, due to the wide range of GSK3 cellular targets, global inhibition of the kinase leads to severe side-effects and GSK3 inhibitors rarely reach Phase-2 clinical trials. Thus, a selective modulation of a specific cellular pool of GSK3 or specific down- or upstream partners of the kinase might provide more efficient anti-neurodegenerative therapies.
Collapse
Affiliation(s)
- Przemysław Duda
- a Department of Molecular Physiology and Neurobiology , University of Wroclaw , Wroclaw , Poland
| | - Janusz Wiśniewski
- a Department of Molecular Physiology and Neurobiology , University of Wroclaw , Wroclaw , Poland
| | - Tomasz Wójtowicz
- a Department of Molecular Physiology and Neurobiology , University of Wroclaw , Wroclaw , Poland
| | - Olga Wójcicka
- a Department of Molecular Physiology and Neurobiology , University of Wroclaw , Wroclaw , Poland
| | - Michał Jaśkiewicz
- a Department of Molecular Physiology and Neurobiology , University of Wroclaw , Wroclaw , Poland
| | - Dominika Drulis-Fajdasz
- a Department of Molecular Physiology and Neurobiology , University of Wroclaw , Wroclaw , Poland
| | - Dariusz Rakus
- a Department of Molecular Physiology and Neurobiology , University of Wroclaw , Wroclaw , Poland
| | - James A McCubrey
- b Department of Microbiology and Immunology , Brody School of Medicine at East Carolina University , Greenville , NC , USA
| | - Agnieszka Gizak
- a Department of Molecular Physiology and Neurobiology , University of Wroclaw , Wroclaw , Poland
| |
Collapse
|
33
|
Khan MT, Deussing J, Tang Y, Illes P. Astrocytic rather than neuronal P2X7 receptors modulate the function of the tri-synaptic network in the rodent hippocampus. Brain Res Bull 2018; 151:164-173. [PMID: 30098388 DOI: 10.1016/j.brainresbull.2018.07.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/21/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
Whole-cell patch clamp recordings demonstrated that in the dentate gyrus (DG) as well as in the CA3 area of mouse hippocampal slices the prototypic P2X7 receptor (R) agonist dibenzoyl-ATP (Bz-ATP) induced inward current responses both in neurons and astrocytes. Whereas the selective P2X7R antagonist A438079 strongly inhibited both neuronal and astrocytic currents, a combination of ionotropic glutamate receptor (CNQX, AP-5) and GABAA-R (gabazine) antagonists depressed the Bz-ATP-induced current responses in the DG (granule cells) and CA3 neurons only. It was concluded that Bz-ATP activated astrocytic P2X7Rs and thereby released glutamate and GABA to stimulate nearby neurons. The residual A438079-resistant current response of astrocytes was suggested to be due to the stimulation of P2XRs of the non-P2X7-type. Further, we searched for presynaptic P2X7Rs at the axon terminals of DG and CA3 pyramidal neurons innervating CA3 and CA1 cells, respectively. Bz-ATP potentiated the frequency of spontaneous postsynaptic currents (sPSCs) in CA1 but not CA3 pyramidal cells. However, the Bz-ATP effect in CA1 cells was inhibited by gabazine or the astrocytic toxin fluorocitrate suggesting stimulation of P2X7Rs at stratum radiatum astrocytes located near to interneurons and synapsing onto CA1 neurons. Our data suggest that functional P2X7Rs are missing at neurons in the tri-synaptic network of the rodent hippocampus, but are present at nearby astrocytes indirectly regulating network activity.
Collapse
Affiliation(s)
- Muhammad Tahir Khan
- Rudolf-Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04107 Leipzig, Germany
| | - Jan Deussing
- Department of Molecular Neurogenetics, Max-Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Peter Illes
- Rudolf-Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04107 Leipzig, Germany; Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
34
|
Roszek K, Wujak M. How to influence the mesenchymal stem cells fate? Emerging role of ectoenzymes metabolizing nucleotides. J Cell Physiol 2018; 234:320-334. [PMID: 30078187 DOI: 10.1002/jcp.26904] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/13/2018] [Indexed: 12/11/2022]
Abstract
Extracellular purines, principally adenosine triphosphate and adenosine, are among the oldest evolutionary and widespread chemical messengers. The integrative view of purinergic signaling as a multistage coordinated cascade involves the participation of nucleotides/nucleosides, their receptors, enzymes metabolizing extracellular nucleosides and nucleotides as well as several membrane transporters taking part in the release and/or uptake of these molecules. In view of the emerging data, it is evident and widely accepted that an extensive network of diverse enzymatic activities exists in the extracellular space. The enzymes regulate the availability of nucleotide and adenosine receptor agonists, and consequently, the course of signaling events. The current data indicate that mesenchymal stem cells (MSCs) and cells induced to differentiate exhibit different sensitivity to purinergic ligands as well as a distinct activity and expression profiles of ectonucleotidases than mature cells. In the proposed review, we postulate for a critical role of these enzymatic players which, by orchestrating a fine-tune regulation of nucleotides concentrations, are integrally involved in modulation and diversification of purinergic signals. This specific hallmark of the MSC purinome should be linked with cell-specific biological potential and capacity for tissue regeneration. We anticipate this publication to be a starting point for scientific discussion and novel approach to the in vitro and in vivo regulation of the MSC properties.
Collapse
Affiliation(s)
- Katarzyna Roszek
- Biochemistry Department, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Magdalena Wujak
- Biochemistry Department, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
35
|
Stefani J, Tschesnokowa O, Parrilla M, Robaye B, Boeynaems JM, Acker-Palmer A, Zimmermann H, Gampe K. Disruption of the Microglial ADP Receptor P2Y 13 Enhances Adult Hippocampal Neurogenesis. Front Cell Neurosci 2018; 12:134. [PMID: 29867367 PMCID: PMC5966569 DOI: 10.3389/fncel.2018.00134] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/30/2018] [Indexed: 12/20/2022] Open
Abstract
In mammalian species, including humans, the hippocampal dentate gyrus (DG) is a primary region of adult neurogenesis. Aberrant adult hippocampal neurogenesis is associated with neurological pathologies. Understanding the cellular mechanisms controlling adult hippocampal neurogenesis is expected to open new therapeutic strategies for mental disorders. Microglia is intimately associated with neural progenitor cells in the hippocampal DG and has been implicated, under varying experimental conditions, in the control of the proliferation, differentiation and survival of neural precursor cells. But the underlying mechanisms remain poorly defined. Using fluorescent in situ hybridization we show that microglia in brain express the ADP-activated P2Y13 receptor under basal conditions and that P2ry13 mRNA is absent from neurons, astrocytes, and neural progenitor cells. Disrupting P2ry13 decreases structural complexity of microglia in the hippocampal subgranular zone (SGZ). But it increases progenitor cell proliferation and new neuron formation. Our data suggest that P2Y13 receptor-activated microglia constitutively attenuate hippocampal neurogenesis. This identifies a signaling pathway whereby microglia, via a nucleotide-mediated mechanism, contribute to the homeostatic control of adult hippocampal neurogenesis. Selective P2Y13R antagonists could boost neurogenesis in pathological conditions associated with impaired hippocampal neurogenesis.
Collapse
Affiliation(s)
- Jennifer Stefani
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, Goethe-University, Frankfurt am Main, Germany
| | - Olga Tschesnokowa
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, Goethe-University, Frankfurt am Main, Germany
| | - Marta Parrilla
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, Goethe-University, Frankfurt am Main, Germany.,Max-Planck-Institute for Brain Research, Frankfurt am Main, Germany
| | - Bernard Robaye
- Institute of Interdisciplinary Research, School of Medicine, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Jean-Marie Boeynaems
- Institute of Interdisciplinary Research, School of Medicine, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, Goethe-University, Frankfurt am Main, Germany.,Max-Planck-Institute for Brain Research, Frankfurt am Main, Germany.,Focus Program Translational Neurosciences (FTN), University of Mainz, Mainz, Germany
| | - Herbert Zimmermann
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, Goethe-University, Frankfurt am Main, Germany
| | - Kristine Gampe
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
36
|
Spinal Cord Stem Cells In Their Microenvironment: The Ependyma as a Stem Cell Niche. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:55-79. [PMID: 29204829 DOI: 10.1007/978-3-319-69194-7_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The ependyma of the spinal cord is currently proposed as a latent neural stem cell niche. This chapter discusses recent knowledge on the developmental origin and nature of the heterogeneous population of cells that compose this stem cell microenviroment, their diverse physiological properties and regulation. The chapter also reviews relevant data on the ependymal cells as a source of plasticity for spinal cord repair.
Collapse
|
37
|
Alves M, Beamer E, Engel T. The Metabotropic Purinergic P2Y Receptor Family as Novel Drug Target in Epilepsy. Front Pharmacol 2018; 9:193. [PMID: 29563872 PMCID: PMC5851315 DOI: 10.3389/fphar.2018.00193] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 02/20/2018] [Indexed: 12/21/2022] Open
Abstract
Epilepsy encompasses a heterogeneous group of neurological syndromes which are characterized by recurrent seizures affecting over 60 million people worldwide. Current anti-epileptic drugs (AEDs) are mainly designed to target ion channels and/or GABA or glutamate receptors. Despite recent advances in drug development, however, pharmacoresistance in epilepsy remains as high as 30%, suggesting the need for the development of new AEDs with a non-classical mechanism of action. Neuroinflammation is increasingly recognized as one of the key players in seizure generation and in the maintenance of the epileptic phenotype. Consequently, targeting signaling molecules involved in inflammatory processes may represent new avenues to improve treatment in epilepsy. Nucleotides such as adenosine-5′-triphosphate (ATP) and uridine-5′-triphosphate (UTP) are released in the brain into the extracellular space during pathological conditions such as increased neuronal firing or cell death. Once released, these nucleotides bind to and activate specific purinergic receptors termed P2 receptors where they mediate the release of gliotransmitters and drive neuronal hyperexcitation and neuroinflammatory processes. This includes the fast acting ionotropic P2X channels and slower-acting G-protein-coupled P2Y receptors. While the expression and function of P2X receptors has been well-established in experimental models of epilepsy, emerging evidence is now also suggesting a prominent role for the P2Y receptor subfamily in seizure generation and the maintenance of epilepsy. In this review we discuss data supporting a role for the P2Y receptor family in epilepsy and the most recent finding demonstrating their involvement during seizure-induced pathology and in epilepsy.
Collapse
Affiliation(s)
- Mariana Alves
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Edward Beamer
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tobias Engel
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
38
|
Wang Y, Huang A, Gan L, Bao Y, Zhu W, Hu Y, Ma L, Wei S, Lan Y. Screening of Potential Genes and Transcription Factors of Postoperative Cognitive Dysfunction via Bioinformatics Methods. Med Sci Monit 2018; 24:503-510. [PMID: 29374768 PMCID: PMC5791419 DOI: 10.12659/msm.907445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The aim of this study was to explore the potential genes and transcription factors involved in postoperative cognitive dysfunction (POCD) via bioinformatics analysis. Material/Methods GSE95070 miRNA expression profiles were downloaded from Gene Expression Omnibus database, which included five hippocampal tissues from POCD mice and controls. Moreover, the differentially expressed miRNAs (DEMs) between the two groups were identified. In addition, the target genes of DEMs were predicted using Targetscan 7.1, followed by protein-protein interaction (PPI) network construction, functional enrichment analysis, pathway analysis, and prediction of transcription factors (TFs) targeting the potential targets. Results A total of eight DEMs were obtained, and 823 target genes were predicted, including 170 POCD-associated genes. Furthermore, potential key genes in the network were remarkably enriched in focal adhesion, protein digestion and absorption, ECM-receptor interaction, and Wnt and MAPK signaling pathways. Conclusions Most potential target genes were involved in the regulation of TFs, including LEF1, SP1, and AP4, which may exert strong impact on the development of POCD.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Anesthesiology, People’s Hospital of Guangxi Zhuang Autonomous
Region, Nanning, Guangxi, P.R. China
| | - Ailan Huang
- Department of Anesthesiology, People’s Hospital of Guangxi Zhuang Autonomous
Region, Nanning, Guangxi, P.R. China
| | - Lixia Gan
- Department of Anesthesiology, People’s Hospital of Guangxi Zhuang Autonomous
Region, Nanning, Guangxi, P.R. China
| | - Yanli Bao
- Department of Anesthesiology, People’s Hospital of Guangxi Zhuang Autonomous
Region, Nanning, Guangxi, P.R. China
| | - Weilin Zhu
- Department of Anesthesiology, People’s Hospital of Guangxi Zhuang Autonomous
Region, Nanning, Guangxi, P.R. China
| | - Yanyan Hu
- Department of Anesthesiology, People’s Hospital of Guangxi Zhuang Autonomous
Region, Nanning, Guangxi, P.R. China
| | - Li Ma
- Department of Anesthesiology, People’s Hospital of Guangxi Zhuang Autonomous
Region, Nanning, Guangxi, P.R. China
| | - Shiyang Wei
- Department of Gynecology, People’s Hospital of Guangxi Zhuang Autonomous
Region, Nanning, Guangxi, P.R. China
| | - Yuyan Lan
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical
University, Nanning, Guangxi, P.R. China
| |
Collapse
|
39
|
Towards a Novel Class of Multitarget-Directed Ligands: Dual P2X7-NMDA Receptor Antagonists. Molecules 2018; 23:molecules23010230. [PMID: 29361735 PMCID: PMC6017257 DOI: 10.3390/molecules23010230] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 01/09/2023] Open
Abstract
Multi-target-directed ligands (MTDLs) offer new hope for the treatment of multifactorial complex diseases such as Alzheimer's Disease (AD). Herein, we present compounds aimed at targeting the NMDA and the P2X7 receptors, which embody a different approach to AD therapy. On one hand, we are seeking to delay neurodegeneration targeting the glutamatergic NMDA receptors; on the other hand, we also aim to reduce neuroinflammation, targeting P2X7 receptors. Although the NMDA receptor is a widely recognized therapeutic target in treating AD, the P2X7 receptor remains largely unexplored for this purpose; therefore, the dual inhibitor presented herein-which is open to further optimization-represents the first member of a new class of MTDLs.
Collapse
|
40
|
Purinergic system in psychiatric diseases. Mol Psychiatry 2018; 23:94-106. [PMID: 28948971 DOI: 10.1038/mp.2017.188] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/15/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
Psychiatric disorders are debilitating diseases, affecting >80 million people worldwide. There are no causal cures for psychiatric disorders and available therapies only treat the symptoms. The etiology of psychiatric disorders is unknown, although it has been speculated to be a combination of environmental, stress and genetic factors. One of the neurotransmitter systems implicated in the biology of psychiatric disorders is the purinergic system. In this review, we performed a comprehensive search of the literature about the role and function of the purinergic system in the development and predisposition to psychiatric disorders, with a focus on depression, schizophrenia, bipolar disorder, autism, anxiety and attention deficit/hyperactivity disorder. We also describe how therapeutics used for psychiatric disorders act on the purinergic system.
Collapse
|
41
|
Menéndez-Méndez A, Díaz-Hernández JI, Ortega F, Gualix J, Gómez-Villafuertes R, Miras-Portugal MT. Specific Temporal Distribution and Subcellular Localization of a Functional Vesicular Nucleotide Transporter (VNUT) in Cerebellar Granule Neurons. Front Pharmacol 2017; 8:951. [PMID: 29311945 PMCID: PMC5744399 DOI: 10.3389/fphar.2017.00951] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/15/2017] [Indexed: 12/14/2022] Open
Abstract
Adenosine triphosphate (ATP) is an important extracellular neurotransmitter that participates in several critical processes like cell differentiation, neuroprotection or axon guidance. Prior to its exocytosis, ATP must be stored in secretory vesicles, a process that is mediated by the Vesicular Nucleotide Transporter (VNUT). This transporter has been identified as the product of the SLC17A9 gene and it is prominently expressed in discrete brain areas, including the cerebellum. The main population of cerebellar neurons, the glutamatergic granule neurons, depends on purinergic signaling to trigger neuroprotective responses. However, while nucleotide receptors like P2X7 and P2Y13 are known to be involved in neuroprotection, the mechanisms that regulate ATP release in relation to such events are less clearly understood. In this work, we demonstrate that cerebellar granule cells express a functional VNUT that is involved in the regulation of ATP exocytosis. Numerous vesicles loaded with this nucleotide can be detected in these granule cells and are staining by the fluorescent ATP-marker, quinacrine. High potassium stimulation reduces quinacrine fluorescence in granule cells, indicating they release ATP via calcium dependent exocytosis. Specific subcellular markers were used to assess the localization of VNUT in granule cells, and the transporter was detected in both the axonal and somatodendritic compartments, most predominantly in the latter. However, co-localization with the specific lysosomal marker LAMP-1 indicated that VNUT can also be found in non-synaptic vesicles, such as lysosomes. Interestingly, the weak co-localization between VNUT and VGLUT1 suggests that the ATP and glutamate vesicle pools are segregated, as also observed in the cerebellar cortex. During post-natal cerebellar development, VNUT is found in granule cell precursors, co-localizing with markers of immature cells like doublecortin, suggesting that this transporter may be implicated in the initial stages of granule cell development.
Collapse
Affiliation(s)
- Aida Menéndez-Méndez
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Complutense University of Madrid, Madrid, Spain.,University Institute of Neurochemistry Research (IUIN), Complutense University of Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Juan I Díaz-Hernández
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Complutense University of Madrid, Madrid, Spain.,University Institute of Neurochemistry Research (IUIN), Complutense University of Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Felipe Ortega
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Complutense University of Madrid, Madrid, Spain.,University Institute of Neurochemistry Research (IUIN), Complutense University of Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Javier Gualix
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Complutense University of Madrid, Madrid, Spain.,University Institute of Neurochemistry Research (IUIN), Complutense University of Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Rosa Gómez-Villafuertes
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Complutense University of Madrid, Madrid, Spain.,University Institute of Neurochemistry Research (IUIN), Complutense University of Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - María T Miras-Portugal
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Complutense University of Madrid, Madrid, Spain.,University Institute of Neurochemistry Research (IUIN), Complutense University of Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
42
|
de Azevedo MI, Da Silva AS, Ferreiro L, Doleski PH, Tonin AA, Casali EA, Moritz CE, Schirmbeck GH, Cardoso VV, Flores MM, Fighera R, Santurio JM. Serum and brain purine levels in an experimental systemic infection of mice by Cryptococcus neoformans : Purinergic immunomodulatory effects. Microb Pathog 2017; 113:124-128. [DOI: 10.1016/j.micpath.2017.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/04/2017] [Accepted: 10/12/2017] [Indexed: 12/20/2022]
|
43
|
P2X7R Blockade Prevents NLRP3 Inflammasome Activation and Pancreatic Fibrosis in a Mouse Model of Chronic Pancreatitis. Pancreas 2017; 46:1327-1335. [PMID: 28930866 DOI: 10.1097/mpa.0000000000000928] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the role of P2X7R (purinergic 2X7 receptor) and NLRP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome activation in the process of pancreatic fibrosis in a mouse model of chronic pancreatitis (CP). METHODS Chronic pancreatitis was induced by repeated intraperitoneal injections of 50 μg/kg cerulein for 6 weeks in mice. P2X7R antagonist oxidized ATP (OxATP) or brilliant blue G (BBG) was administered after the last cerulein injection for 2 weeks. Pancreatic chronic inflammation and fibrosis were evaluated by histological score, Sirius red staining, and alpha-smooth muscle actin immunohistochemical staining. We further determined pancreatic P2X7R, NLRP3, and caspase-1 expressions in gene and protein levels and the pancreatic concentrations of caspase-1, interleukin 1β (IL-1β), and IL-18. RESULTS The pancreatic P2X7R, NLRP3, and caspase-1 expressions in gene and protein levels and the pancreatic concentrations of caspase-1, IL-1β, and IL-18 were all reduced significantly in both the OxATP and BBG groups (P < 0.05). The pancreatic chronic inflammation and the fibrosis indices were all remarkably attenuated (P < 0.05). CONCLUSIONS P2X7R antagonist OxATP and BBG significantly decreased pancreatic chronic inflammation and fibrosis in a mouse CP model and suggested that blockade of P2X7R-NLRP3 inflammasome signaling pathway may represent a novel therapeutic strategy for CP and its fibrotic process.
Collapse
|
44
|
Olfactory ensheathing cell transplantation inhibits P2X4 receptor overexpression in spinal cord injury rats with neuropathic pain. Neurosci Lett 2017; 651:171-176. [DOI: 10.1016/j.neulet.2017.04.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/11/2017] [Accepted: 04/27/2017] [Indexed: 12/13/2022]
|
45
|
P2X7 Participates in Intracerebral Hemorrhage-Induced Secondary Brain Injury in Rats via MAPKs Signaling Pathways. Neurochem Res 2017; 42:2372-2383. [PMID: 28488233 DOI: 10.1007/s11064-017-2257-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 01/31/2023]
Abstract
This study aimed to study the role of P2X7 in intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI) and the underlying mechanisms. An autologous blood injection was used to induce ICH model in Sprague-Dawley rats, and cultured primary rat cortical neurons were exposed to oxyhemoglobin to mimic ICH in vitro. siRNA interference and over-expression of P2X7, agonists and antagonists of P2X7, p38 MAPK and ERK were exploited. The protein levels were assessed using Western blotting and immunofluorescence staining. Terminal deoxynucleotidyl transferase dUTP nick end labeling staining and Fluoro-Jade B were conducted to detect apoptotic and degenerating neurons. The protein levels of P2X7, phosphorylated p38, ERK, active caspase-3 and NF-κB were significantly increased by ICH, which could be further increased by BzATP (P2X7 agonist) and reduced by BBG (P2X7 antagonist). And BzATP demonstrated a significant increase in cell death ratio and brain water content, while BBG led to a reverse results. In addition, Over- P2X7 increased the levels of P2X7, phosphorylated p38, ERK, active caspase-3 and NF-κB, and aggravated cell apoptosis, while si P2X7 resulted in opposite effects. Finally, the protein levels of phosphorylated P38 and active caspase 3 were decreased by BzATP plus Hydrochloride (p38 MAPK antagonist) and increased vy BBG plus Asiatic acid (p38 MAPK agonist), while the protein levels of phosphorylated ERK and NF-κB were decreased with BzATP plus Nimbolide (ERK antagonist) and increased with BBG plus Saikosaponin C (ERK agonist). This study demonstrates that inhibition of P2X7 could prevent ICH-induced SBI via MAPKs signaling pathway.
Collapse
|
46
|
Reigada D, Navarro-Ruiz RM, Caballero-López MJ, Del Águila Á, Muñoz-Galdeano T, Maza RM, Nieto-Díaz M. Diadenosine tetraphosphate (Ap 4A) inhibits ATP-induced excitotoxicity: a neuroprotective strategy for traumatic spinal cord injury treatment. Purinergic Signal 2017; 13:75-87. [PMID: 27761681 PMCID: PMC5334201 DOI: 10.1007/s11302-016-9541-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/27/2016] [Indexed: 01/02/2023] Open
Abstract
Reducing cell death during the secondary injury is a major priority in the development of a cure for traumatic spinal cord injury (SCI). One of the earliest processes that follow SCI is the excitotoxicity resulting from the massive release of excitotoxicity mediators, including ATP, which induce an excessive and/or prolonged activation of their receptors and a deregulation of the calcium homeostasis. Diadenosine tetraphosphate (Ap4A) is an endogenous purinergic agonist, present in both extracellular and intracellular fluids, with promising cytoprotective effects in different diseases including neurodegenerative processes. In a search for efficient neuroprotective strategies for SCI, we have tested the capability of Ap4A to reduce the excitotoxic death mediated by the ATP-induced deregulation of calcium homeostasis and its consequences on tissue preservation and functional recovery in a mouse model of moderate contusive SCI. Our analyses with the murine neural cell line Neuro2a demonstrate that treatment with Ap4A reduces ATP-dependent excitotoxic death by both lowering the intracellular calcium response and decreasing the expression of specific purinergic receptors. Follow-up analyses in a mouse model of contusive SCI showed that acute administration of Ap4A following SCI reduces tissue damage and improves motor function recovery. These results suggest that Ap4A cytoprotection results from a decrease of the purinergic tone preventing the effects of a massive release of ATP after SCI, probably together with a direct induction of anti-apoptotic and pro-survival pathways via activation of P2Y2 proposed in previous studies. In conclusion, Ap4A may be a good candidate for an SCI therapy, particularly to reduce excitotoxicity in combination with other modulators and/or inhibitors of the excitotoxic process that are being tested.
Collapse
Affiliation(s)
- David Reigada
- Molecular Neuroprotection Group, Experimental Neurology Unit, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071 Toledo, Spain
| | - Rosa María Navarro-Ruiz
- Molecular Neuroprotection Group, Experimental Neurology Unit, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071 Toledo, Spain
| | - Marcos Javier Caballero-López
- Molecular Neuroprotection Group, Experimental Neurology Unit, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071 Toledo, Spain
| | - Ángela Del Águila
- Molecular Neuroprotection Group, Experimental Neurology Unit, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071 Toledo, Spain
| | - Teresa Muñoz-Galdeano
- Molecular Neuroprotection Group, Experimental Neurology Unit, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071 Toledo, Spain
| | - Rodrigo M. Maza
- Molecular Neuroprotection Group, Experimental Neurology Unit, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071 Toledo, Spain
| | - Manuel Nieto-Díaz
- Molecular Neuroprotection Group, Experimental Neurology Unit, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071 Toledo, Spain
| |
Collapse
|
47
|
Paniagua-Herranz L, Gil-Redondo JC, Queipo MJ, González-Ramos S, Boscá L, Pérez-Sen R, Miras-Portugal MT, Delicado EG. Prostaglandin E 2 Impairs P2Y 2/P2Y 4 Receptor Signaling in Cerebellar Astrocytes via EP3 Receptors. Front Pharmacol 2017; 8:937. [PMID: 29311938 PMCID: PMC5743739 DOI: 10.3389/fphar.2017.00937] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/11/2017] [Indexed: 02/05/2023] Open
Abstract
Prostaglandin E2 (PGE2) is an important bioactive lipid that accumulates after tissue damage or inflammation due to the rapid expression of cyclooxygenase 2. PGE2 activates specific G-protein coupled EP receptors and it mediates pro- or anti-inflammatory actions depending on the cell-context. Nucleotides can also be released in these situations and they even contribute to PGE2 production. We previously described the selective impairment of P2Y nucleotide signaling by PGE2 in macrophages and fibroblasts, an effect independent of prostaglandin receptors but that involved protein kinase C (PKC) and protein kinase D (PKD) activation. Considering that macrophages and fibroblasts influence inflammatory responses and tissue remodeling, a similar mechanism involving P2Y signaling could occur in astrocytes in response to neuroinflammation and brain repair. We analyzed here the modulation of cellular responses involving P2Y2/P2Y4 receptors by PGE2 in rat cerebellar astrocytes. We demonstrate that PGE2 inhibits intracellular calcium responses elicited by UTP in individual cells and that inhibiting this P2Y signaling impairs the astrocyte migration elicited by this nucleotide. Activation of EP3 receptors by PGE2 not only impairs the calcium responses but also, the extracellular regulated kinases (ERK) and Akt phosphorylation induced by UTP. However, PGE2 requires epidermal growth factor receptor (EGFR) transactivation in order to dampen P2Y signaling. In addition, these effects of PGE2 also occur in a pro-inflammatory context, as evident in astrocytes stimulated with bacterial lipopolysaccharide (LPS). While we continue to investigate the intracellular mechanisms responsible for the inhibition of UTP responses, the involvement of novel PKC and PKD in cerebellar astrocytes cannot be excluded, kinases that could promote the internalization of P2Y receptors in fibroblasts.
Collapse
|
48
|
Mueller M, Oppliger B, Joerger-Messerli M, Reinhart U, Barnea E, Paidas M, Kramer BW, Surbek DV, Schoeberlein A. Wharton's Jelly Mesenchymal Stem Cells Protect the Immature Brain in Rats and Modulate Cell Fate. Stem Cells Dev 2016; 26:239-248. [PMID: 27842457 DOI: 10.1089/scd.2016.0108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The development of a mammalian brain is a complex and long-lasting process. Not surprisingly, preterm birth is the leading cause of death in newborns and children. Advances in perinatal care reduced mortality, but morbidity still represents a major burden. New therapeutic approaches are thus desperately needed. Given that mesenchymal stem/stromal cells (MSCs) emerged as a promising candidate for cell therapy, we transplanted MSCs derived from the Wharton's Jelly (WJ-MSCs) to reduce the burden of immature brain injury in a murine animal model. WJ-MSCs transplantation resulted in protective activity characterized by reduced myelin loss and astroglial activation. WJ-MSCs improved locomotor behavior as well. To address the underlying mechanisms, we tested the key regulators of responses to DNA-damaging agents, such as cyclic AMP-dependent protein kinase/calcium-dependent protein kinase (PKA/PKC), cyclin-dependent kinase (CDK), ataxia-telangiectasia-mutated/ATM- and Rad3-related (ATM/ATR) substrates, protein kinase B (Akt), and 14-3-3 binding protein partners. We characterized WJ-MSCs using a specific profiler polymerase chain reaction array. We provide evidence that WJ-MSCs target pivotal regulators of the cell fate such as CDK/14-3-3/Akt signaling. We identified leukemia inhibitory factor as a potential candidate of WJ-MSCs' induced modifications as well. We hypothesize that WJ-MSCs may exert adaptive responses depending on the type of injury they are facing, making them prominent candidates for cell therapy in perinatal injuries.
Collapse
Affiliation(s)
- Martin Mueller
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland .,3 Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine , New Haven, Connecticut
| | - Byron Oppliger
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| | - Marianne Joerger-Messerli
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| | - Ursula Reinhart
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| | - Eytan Barnea
- 4 Society for the Investigation of Early Pregnancy and BioIncept LLC , Cherry Hill, New Jersey
| | - Michael Paidas
- 3 Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine , New Haven, Connecticut
| | - Boris W Kramer
- 5 Department of Pediatrics, Maastricht University Medical Center (MUMC) , Maastricht, the Netherlands .,6 Division Neuroscience, Department of Neuropsychology, School of Mental Health and Neuroscience (MHeNS), Maastricht University , Maastricht, the Netherlands
| | - Daniel V Surbek
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| | - Andreina Schoeberlein
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| |
Collapse
|
49
|
Engel T, Brennan GP, Sanz-Rodriguez A, Alves M, Beamer E, Watters O, Henshall DC, Jimenez-Mateos EM. A calcium-sensitive feed-forward loop regulating the expression of the ATP-gated purinergic P2X7 receptor via specificity protein 1 and microRNA-22. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:255-266. [PMID: 27840225 DOI: 10.1016/j.bbamcr.2016.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/05/2016] [Accepted: 11/08/2016] [Indexed: 11/26/2022]
Abstract
Cells have developed complex transcriptional regulatory mechanisms to maintain intracellular homeostasis and withstand pathophysiological stressors. Feed-forward loops comprising transcription factors that drive expression of both target gene and a microRNA as negative regulator, are gaining increasing recognition as key regulatory elements of cellular homeostasis. The ATP-gated purinergic P2X7 receptor (P2X7R) is an important driver of inflammation and has been implicated in the pathogenesis of numerous brain diseases including epilepsy. Changes in P2X7R expression have been reported in both experimental models and in epilepsy patients but the mechanism(s) controlling P2X7R levels remain incompletely understood. The specificity protein 1 (Sp1) has been shown to induce P2X7R transcription in vitro and recent data has identified microRNA-22 as a post-transcriptional repressor of P2X7R expression after seizures. In the present study we show that Sp1 can induce the transcription of both microRNA-22 and P2X7R in vitro during increased neuronal activity and in vivo in a mouse model of status epilepticus. We further show that Sp1-driven microRNA-22 transcription is calcium-sensitive and Sp1 occupancy of the microRNA-22 promoter region is blocked under conditions of seizure activity sufficient to elicit neuronal death. Taken together, our results suggest a neuronal activity-dependent P2X7R expression which is induced by the transcription factor Sp1 and repressed in a calcium-dependent manner by microRNA-22.
Collapse
Affiliation(s)
- Tobias Engel
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| | - Gary P Brennan
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Amaya Sanz-Rodriguez
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Mariana Alves
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Edward Beamer
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Orla Watters
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Eva M Jimenez-Mateos
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
50
|
Tang Y, Illes P. Regulation of adult neural progenitor cell functions by purinergic signaling. Glia 2016; 65:213-230. [PMID: 27629990 DOI: 10.1002/glia.23056] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/20/2016] [Accepted: 08/23/2016] [Indexed: 01/30/2023]
Abstract
Extracellular purines are signaling molecules in the neurogenic niches of the brain and spinal cord, where they activate cell surface purinoceptors at embryonic neural stem cells (NSCs) and adult neural progenitor cells (NPCs). Although mRNA and protein are expressed at NSCs/NPCs for almost all subtypes of the nucleotide-sensitive P2X/P2Y, and the nucleoside-sensitive adenosine receptors, only a few of those have acquired functional significance. ATP is sequentially degraded by ecto-nucleotidases to ADP, AMP, and adenosine with agonistic properties for distinct receptor-classes. Nucleotides/nucleosides facilitate or inhibit NSC/NPC proliferation, migration and differentiation. The most ubiquitous effect of all agonists (especially of ATP and ADP) appears to be the facilitation of cell proliferation, usually through P2Y1Rs and sometimes through P2X7Rs. However, usually P2X7R activation causes necrosis/apoptosis of NPCs. Differentiation can be initiated by P2Y2R-activation or P2X7R-blockade. A key element in the transduction mechanism of either receptor is the increase of the intracellular free Ca2+ concentration, which may arise due to its release from intracellular storage sites (G protein-coupling; P2Y) or due to its passage through the receptor-channel itself from the extracellular space (ATP-gated ion channel; P2X). Further research is needed to clarify how purinergic signaling controls NSC/NPC fate and how the balance between the quiescent and activated states is established with fine and dynamic regulation. GLIA 2017;65:213-230.
Collapse
Affiliation(s)
- Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Peter Illes
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, 04107, Germany
| |
Collapse
|