1
|
Karger G, Willker JE, Harders AR, Watermann P, Dringen R. ATP Restoration by ATP-Deprived Cultured Primary Astrocytes. Neurochem Res 2024; 50:13. [PMID: 39549173 PMCID: PMC11569012 DOI: 10.1007/s11064-024-04276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 11/18/2024]
Abstract
A high cellular concentration of adenosine triphosphate (ATP) is essential to fuel many important functions of brain astrocytes. Although cellular ATP depletion has frequently been reported for astrocytes, little is known on the metabolic pathways that contribute to ATP restoration by ATP-depleted astrocytes. Incubation of cultured primary rat astrocytes in glucose-free buffer for 60 min with the mitochondrial uncoupler BAM15 lowered the cellular ATP content by around 70%, the total amount of adenosine phosphates by around 50% and the adenylate energy charge (AEC) from 0.9 to 0.6. Testing for ATP restoration after removal of the uncoupler revealed that the presence of glucose as exclusive substrate allowed the cells to restore within 6 h around 80% of the initial ATP content, while coapplication of adenosine plus glucose enabled the cells to fully restore their initial ATP content within 60 min. A rapid but incomplete and transient ATP restoration was found for astrocytes that had been exposed to adenosine alone. This restoration was completely prevented by application of the pyruvate uptake inhibitor UK5099, the respiratory chain inhibitor antimycin A or by the continuous presence of BAM15. However, the presence of these compounds strongly accelerated the release of lactate from the cells, suggesting that the ribose moiety of adenosine can serve as substrate to fuel some ATP restoration via mitochondrial metabolism. Finally, the adenosine-accelerated ATP restoration in glucose-fed astrocytes was inhibited by the presence of the adenosine kinase inhibitor ABT-702. These data demonstrate that astrocytes require for a rapid and complete ATP restoration the presence of both glucose as substrate and adenosine as AMP precursor.
Collapse
Affiliation(s)
- Gabriele Karger
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Johanna Elisabeth Willker
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Antonia Regina Harders
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Patrick Watermann
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany.
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany.
| |
Collapse
|
2
|
FAN H, GE X, ZHOU X, LI Y, LIU Q, HU Y. [Non-targeted Metabolomics-based Exploration of
Radiation-induced Metabolic Alterations in Mouse Lung Epithelial Cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:725-734. [PMID: 39631829 PMCID: PMC11629006 DOI: 10.3779/j.issn.1009-3419.2024.106.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Metabolic change is one of the important characteristics of radiation pneumonitis. Radiotherapy, as a conventional method for the treatment of thoracic tumors, can not only effectively kill tumor cells, but also cause adverse reactions such as local inflammation and fibrosis, which leads to limited therapeutic effect and profound impact on the quality of life of patients. Therefore, it is of great significance to explore the metabolic changes caused by radiotherapy. The aim of this study was to investigate the effects of X-ray irradiation on the metabolism of a mouse lung epithelial cell line (murine lung epithelial-12, MLE12). METHODS MLE12 cells were' cultured in vitro and randomly divided into radiation group (IR) and control group (NC). Cells in the IR group were irradiated at a dose of 10 Gy using a Hitachi X-ray irradiator. Cell supernatant samples were collected at 48 h after irradiation. Metabolomic analysis of the samples was performed by liquid chromatograph mass spectrometer (LC/MS). RESULTS LC/MS metabolomics analysis revealed the metabolic changes of MLE12 cells at 48 h after irradiation. A total of 38 secretory metabolites were altered in the IR group compared with the NC group. According to the annotation of Kyoto Encyclopedia of Genes and Genomes (KEGG) database, the differential metabolites are mainly involved in nucleotide metabolism, amino acid metabolism and lipid metabolism, among which the difference in nucleotide metabolism is the most significant. CONCLUSIONS The metabolism of MLE12 cells was significantly affected by X-ray irradiation, mainly affecting the nucleotide metabolic pathways, including purine and pyrimidine metabolites and related metabolic pathways.
Collapse
|
3
|
Qu Z, Tian P, Wang L, Jin X, Guo M, Lu J, Zhao J, Chen W, Wang G. Dietary Nucleotides Promote Neonatal Rat Microbiota-Gut-Brain Axis Development by Affecting Gut Microbiota Composition and Metabolic Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:19622-19637. [PMID: 38014964 DOI: 10.1021/acs.jafc.3c07349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
A variety of active factors in milk and foods have been proven to serve as microbial nutrients that regulate the formation of early gut microbiota (GM), thereby ensuring the healthy development of infants. This study demonstrated that dietary nucleotides (NTs), one of the main nitrogen-containing substances in human milk, promoted the neurodevelopment of neonatal rats and the expression of Sox2, Dcx, Tuj1, and NeuN in the prefrontal cortex and hippocampus, but had no significant regulatory effects in the striatum. 16s rRNA sequencing and metabolomics of the colon contents of neonatal rats at different developmental stages showed that the early intake of NTs promoted an increase in the abundance of beneficial microorganisms related to neurodevelopment, digestion, and gut absorption, such as g_Romboutsia and g_Akkermansia. Changes in the ability of the GM to regulate folate synthesis, riboflavin metabolism, and other processes were also observed. Further analysis revealed significant correlations between the level of characteristic metabolites, namely, trans-3-indoleacrylic acid, urocanic acid, inosine, and adenosine, in the gut with neurodevelopment and characteristic GM components. These findings suggest that NTs in milk may affect neurodevelopment and maturation in early life by regulating the GM composition-gut-brain axis.
Collapse
Affiliation(s)
- Zhihao Qu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Linlin Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xing Jin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Min Guo
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jingyu Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| |
Collapse
|
4
|
Ölmez A, Çetin GO, Karaer K. Early onset disease, anarthria, areflexia, and dystonia can be the distinctive features of SPG64, a very rare form of hereditary spastic paraplegias. Am J Med Genet A 2022; 188:2712-2717. [PMID: 35758610 DOI: 10.1002/ajmg.a.62878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 05/22/2022] [Accepted: 06/02/2022] [Indexed: 01/25/2023]
Abstract
Hereditary spastic paraplegias (HSP) are a group of inherited, neurodegenerative disorders characterized by progressive gait impairment, lower extremity spasticity and increased patellar reflexes. More than 80 types of HSP have been defined to date. In complicated forms, lower limb spasticity and gait impairment is accompanied by an additional neurological finding. Autosomal recessive (AR) HSPs are usually identified in complicated forms and occur more frequently in countries where consanguineous marriage is more widespread. Next generation sequencing techniques, developed in the last decade, have led to the identification of many new types of HSP and reduced the "diagnostic odyssey." Whole exome sequencing (WES) can diagnose up to 75% of undiagnosed HSP patients. Targeted genetic analysis with good clinical phenotyping gives the best diagnostic yields for rare diseases. Clinical heterogeneity is prominent in AR complicated HSP. However, some clinical features complicating the disease or magnetic resonance imaging findings, including thin corpus callosum or white matter abnormalities, can help to distinguish some types. AR spastic paraplegia type 64 (SPG64) is a very rare HSP, caused by a mutation in the ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1) gene, first described in 2014. To date only nine patients from five families have been reported. We present two siblings with a novel pathogenic variant in ENTPD1, diagnosed by WES, as the sixth published family. We propose that early onset in childhood, cognitive impairment, dysarthria/anarthria, dystonia and areflexia may be the distinctive features of SPG64 and more clinical evidence from families with pathogenic ENTPD1 variants is warranted.
Collapse
Affiliation(s)
- Akgün Ölmez
- Pediatric Neurology Private Office, Denizli, Turkey
| | - Gökhan Ozan Çetin
- Faculty Of Medicine, Department of Medical Genetics, Pamukkale University, Denizli, Turkey
| | - Kadri Karaer
- Faculty Of Medicine, Department of Medical Genetics, Pamukkale University, Denizli, Turkey
| |
Collapse
|
5
|
Wang X, Cui L, Ji X. Cognitive impairment caused by hypoxia: from clinical evidences to molecular mechanisms. Metab Brain Dis 2022; 37:51-66. [PMID: 34618295 DOI: 10.1007/s11011-021-00796-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/09/2021] [Indexed: 12/23/2022]
Abstract
Hypoxia is a state of reduced oxygen supply and excessive oxygen consumption. According to the duration of hypoxic period, it can be classified as acute and chronic hypoxia. Both acute and chronic hypoxia could induce abundant neurological deficits. Although there have been significant advances in the pathophysiological injuries, few studies have focused on the cognitive dysfunction. In this review, we focused on the clinical evidences and molecular mechanisms of cognitive impairment under acute and chronic hypoxia. Hypoxia can impair several cognitive domains such as attention, learning and memory, procession speed and executive function, which are similar in acute and chronic hypoxia. The severity of cognitive deficit correlates with the duration and degree of hypoxia. Recovery can be achieved after acute hypoxia, while sequelae or even dementia can be observed after chronic hypoxia, perhaps due to the different molecular mechanisms. Cardiopulmonary compensatory response, glycolysis, oxidative stress, calcium overload, adenosine, mitochondrial disruption, inflammation and excitotoxicity contribute to the molecular mechanisms of cognitive deficit after acute hypoxia. During the chronic stage of hypoxia, different adaptive responses, impaired neurovascular coupling, apoptosis, transcription factors-mediated inflammation, as well as Aβ accumulation and tau phosphorylation account for the neurocognitive deficit. Moreover, brain structural changes with hippocampus and cortex atrophy, ventricle enlargement, senile plaque and neurofibrillary tangle deposition can be observed under chronic hypoxia rather than acute hypoxia.
Collapse
Affiliation(s)
- Xiaoyin Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Lili Cui
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, No 45, Changchun Street, Beijing, 100053, Xicheng District, China.
| |
Collapse
|
6
|
Beamer E, O’Dea MI, Garvey AA, Smith J, Menéndez-Méndez A, Kelly L, Pavel A, Quinlan S, Alves M, Jimenez-Mateos EM, Tian F, Dempsey E, Dale N, Murray DM, Boylan GB, Molloy EJ, Engel T. Novel Point-of-Care Diagnostic Method for Neonatal Encephalopathy Using Purine Nucleosides. Front Mol Neurosci 2021; 14:732199. [PMID: 34566578 PMCID: PMC8458851 DOI: 10.3389/fnmol.2021.732199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/19/2021] [Indexed: 01/19/2023] Open
Abstract
Background: Evidence suggests that earlier diagnosis and initiation of treatment immediately after birth is critical for improved neurodevelopmental outcomes following neonatal encephalopathy (NE). Current diagnostic tests are, however, mainly restricted to clinical diagnosis with no molecular tests available. Purines including adenosine are released during brain injury such as hypoxia and are also present in biofluids. Whether blood purine changes can be used to diagnose NE has not been investigated to date. Methods: Blood purines were measured in a mouse model of neonatal hypoxia and infants with NE using a novel point-of-care diagnostic technology (SMARTChip) based on the summated electrochemical detection of adenosine and adenosine metabolites in the blood. Results: Blood purine concentrations were ∼2-3-fold elevated following hypoxia in mice [2.77 ± 0.48 μM (Control) vs. 7.57 ± 1.41 μM (post-hypoxia), p = 0.029]. Data in infants with NE had a 2-3-fold elevation when compared to healthy controls [1.63 ± 0.47 μM (Control, N = 5) vs. 4.87 ± 0.92 μM (NE, N = 21), p = 0.0155]. ROC curve analysis demonstrates a high sensitivity (81%) and specificity (80%) for our approach to identify infants with NE. Moreover, blood purine concentrations were higher in infants with NE and seizures [8.13 ± 3.23 μM (with seizures, N = 5) vs. 3.86 ± 0.56 μM (without seizures, N = 16), p = 0.044]. Conclusion: Our data provides the proof-of-concept that measurement of blood purine concentrations via SMARTChip technology may offer a low-volume bedside test to support a rapid diagnosis of NE.
Collapse
Affiliation(s)
- Edward Beamer
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
- Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Mary Isabel O’Dea
- Coombe Women and Infants University Hospital, Dublin, Ireland
- National Children’s Research Centre, Crumlin, Dublin, Ireland
- Discipline of Paediatrics, Children’s Health Ireland at Crumlin and Tallaght, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Aisling A. Garvey
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Jonathon Smith
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Aida Menéndez-Méndez
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Lynne Kelly
- Coombe Women and Infants University Hospital, Dublin, Ireland
- Discipline of Paediatrics, Children’s Health Ireland at Crumlin and Tallaght, Dublin, Ireland
| | - Andreea Pavel
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Sean Quinlan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Mariana Alves
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Eva M. Jimenez-Mateos
- Discipline of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Faming Tian
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Eugene Dempsey
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Deirdre M. Murray
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Geraldine B. Boylan
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Eleanor J. Molloy
- Coombe Women and Infants University Hospital, Dublin, Ireland
- National Children’s Research Centre, Crumlin, Dublin, Ireland
- Discipline of Paediatrics, Children’s Health Ireland at Crumlin and Tallaght, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
7
|
Pashaei M, Davarzani A, Hajati R, Zamani B, Nafissi S, Larti F, Nilipour Y, Rohani M, Alavi A. Description of clinical features and genetic analysis of one ultra-rare (SPG64) and two common forms (SPG5A and SPG15) of hereditary spastic paraplegia families. J Neurogenet 2021; 35:84-94. [PMID: 33771085 DOI: 10.1080/01677063.2021.1895146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hereditary spastic paraplegia (HSP) is a clinically and genetically heterogeneous neurodegenerative disorder, characterized by lower-limb spasticity and weakness. To date, more than 82 loci/genes (SPG1-SPG82) have been identified that contribute to the cause of HSP. Despite the use of next-generation sequencing-based methods, genetic-analysis has failed in the finding of causative genes in more than 50% of HSP patients, indicating a more significant heterogeneity and absence of a given phenotype-genotype correlation. Here, we performed whole-exome sequencing (WES) to identify HSP-causing genes in three unrelated-Iranian probands. Candidate variants were detected and confirmed in the probands and co-segregated in the family members. The phenotypic data gathered and compared with earlier cases with the same sub-types of disease. Three novel homozygous variants, c.978delT; p.Q327Kfs*39, c.A1208G; p.D403G and c.3811delT; p.S1271Lfs*44, in known HSP-causing genes including ENTPD1, CYP7B1, and ZFYVE26 were identified, respectively. Intra and interfamilial clinical variability were observed among affected individuals. Mutations in CYP7B1 and ZFYVE26 are relatively common causes of HSP and associated with SPG5A and SPG15, respectively. However, mutations in ENTPD1 are related to SPG64 which is an ultra-rare form of HSP. The research affirmed more complexities of phenotypic manifestations and allelic heterogeneity in HSP. Due to these complexities, it is not feasible to show a clear phenotype-genotype correlation in HSP cases. Identification of more families with mutations in HSP-causing genes may help the establishment of this correlation, further understanding of the molecular basis of the disease, and would provide an opportunity for genetic-counseling in these families.
Collapse
Affiliation(s)
- Mahdieh Pashaei
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Atefeh Davarzani
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Reza Hajati
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Babak Zamani
- Neurology Department, Firoozgar hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Shahriar Nafissi
- Department of Neurology, Shariati Hospital., Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Larti
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Yalda Nilipour
- Pediatric Pathology Research Center, Research Institute for Children Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rohani
- Department of Neurology, Hazrat Rasool Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Afagh Alavi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| |
Collapse
|
8
|
Adenosine Signaling and Clathrin-Mediated Endocytosis of Glutamate AMPA Receptors in Delayed Hypoxic Injury in Rat Hippocampus: Role of Casein Kinase 2. Mol Neurobiol 2021; 58:1932-1951. [PMID: 33415682 PMCID: PMC8018935 DOI: 10.1007/s12035-020-02246-0] [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: 08/29/2019] [Accepted: 12/03/2020] [Indexed: 11/20/2022]
Abstract
Chronic adenosine A1R stimulation in hypoxia leads to persistent hippocampal synaptic depression, while unopposed adenosine A2AR receptor stimulation during hypoxia/reperfusion triggers adenosine-induced post-hypoxia synaptic potentiation (APSP) and increased neuronal death. Still, the mechanisms responsible for this adenosine-mediated neuronal damage following hypoxia need to be fully elucidated. We tested the hypothesis that A1R and A2AR regulation by protein kinase casein kinase 2 (CK2) and clathrin-dependent endocytosis of AMPARs both contribute to APSPs and neuronal damage. The APSPs following a 20-min hypoxia recorded from CA1 layer of rat hippocampal slices were abolished by A1R and A2AR antagonists and by broad-spectrum AMPAR antagonists. The inhibitor of GluA2 clathrin-mediated endocytosis Tat-GluA2-3Y peptide and the dynamin-dependent endocytosis inhibitor dynasore both significantly inhibited APSPs. The CK2 antagonist DRB also inhibited APSPs and, like hypoxic treatment, caused opposite regulation of A1R and A2AR surface expression. APSPs were abolished when calcium-permeable AMPAR (CP-AMPAR) antagonist (IEM or philanthotoxin) or non-competitive AMPAR antagonist perampanel was applied 5 min after hypoxia. In contrast, perampanel, but not CP-AMPAR antagonists, abolished APSPs when applied during hypoxia/reperfusion. To test for neuronal viability after hypoxia, propidium iodide staining revealed significant neuroprotection of hippocampal CA1 pyramidal neurons when pretreated with Tat-GluA2-3Y peptide, CK2 inhibitors, dynamin inhibitor, CP-AMPAR antagonists (applied 5 min after hypoxia), and perampanel (either at 5 min hypoxia onset or during APSP). These results suggest that the A1R-CK2-A2AR signaling pathway in hypoxia/reperfusion injury model mediates increased hippocampal synaptic transmission and neuronal damage via calcium-permeable AMPARs that can be targeted by perampanel for neuroprotective stroke therapy.
Collapse
|
9
|
Townsend MH, Tellez Freitas CM, Larsen D, Piccolo SR, Weber KS, Robison RA, O'Neill KL. Hypoxanthine Guanine Phosphoribosyltransferase expression is negatively correlated with immune activity through its regulation of purine synthesis. Immunobiology 2020; 225:151931. [PMID: 32291109 DOI: 10.1016/j.imbio.2020.151931] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/13/2020] [Accepted: 03/03/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The purpose of this study was to examine the effects of elevated Hypoxanthine Guanine Phosphoribosyltransferase (HPRT) on the immune response in the tumor microenvironment. METHODOLOGY HPRT expression was evaluated in cancer patients and correlated with cytokine expression, survival, and immune cell infiltration. An HPRT knockdown cell line was created to evaluate HPRT impact on purine expression and subsequent purine treatment was administered to immune cells to determine their influence on cell activation. RESULTS HPRT expression was negatively correlated with the general expression of both pro-inflammatory and anti-inflammatory cytokines. Additionally, HPRT expression was also negatively correlated with the infiltration of immune cell subsets: B-cells, CD4 + T cells, macrophages, neutrophils, and dendritic cells (p < 0.001) and CD8 + T-cells (p < 0.01). When HPRT was knocked down in a Raji cell line, the levels of adenosine were reduced significantly compared to the wild type. When examining the level of Ca2+ influx of Raji compared to the HPRT Raji knockdown cell, there was a significant decrease in calcium influx in the knockdown cells when compared to the wild type cells. This demonstrates that HPRT had a significant impact on overall cell activation and the ability of the cells to properly influx calcium needed for their activation. CONCLUSIONS We conclude that purine levels significantly reduce immune cell activation in cancer and the upregulation of HPRT in malignant tissue is a contributing factors to the immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Michelle H Townsend
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA.
| | - Claudia M Tellez Freitas
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA; College of Dental Medicine, Roseman University of Health Science, South Jordan, UT, USA
| | - Dallas Larsen
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Stephen R Piccolo
- Department of Biology, Brigham Young University, Provo, UT, USA; Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, USA
| | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Richard A Robison
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Kim L O'Neill
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| |
Collapse
|
10
|
Reklow RJ, Alvares TS, Zhang Y, Miranda Tapia AP, Biancardi V, Katzell AK, Frangos SM, Hansen MA, Toohey AW, Cass CE, Young JD, Pagliardini S, Boison D, Funk GD. The Purinome and the preBötzinger Complex - A Ménage of Unexplored Mechanisms That May Modulate/Shape the Hypoxic Ventilatory Response. Front Cell Neurosci 2019; 13:365. [PMID: 31496935 PMCID: PMC6712068 DOI: 10.3389/fncel.2019.00365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022] Open
Abstract
Exploration of purinergic signaling in brainstem homeostatic control processes is challenging the traditional view that the biphasic hypoxic ventilatory response, which comprises a rapid initial increase in breathing followed by a slower secondary depression, reflects the interaction between peripheral chemoreceptor-mediated excitation and central inhibition. While controversial, accumulating evidence supports that in addition to peripheral excitation, interactions between central excitatory and inhibitory purinergic mechanisms shape this key homeostatic reflex. The objective of this review is to present our working model of how purinergic signaling modulates the glutamatergic inspiratory synapse in the preBötzinger Complex (key site of inspiratory rhythm generation) to shape the hypoxic ventilatory response. It is based on the perspective that has emerged from decades of analysis of glutamatergic synapses in the hippocampus, where the actions of extracellular ATP are determined by a complex signaling system, the purinome. The purinome involves not only the actions of ATP and adenosine at P2 and P1 receptors, respectively, but diverse families of enzymes and transporters that collectively determine the rate of ATP degradation, adenosine accumulation and adenosine clearance. We summarize current knowledge of the roles played by these different purinergic elements in the hypoxic ventilatory response, often drawing on examples from other brain regions, and look ahead to many unanswered questions and remaining challenges.
Collapse
Affiliation(s)
- Robert J. Reklow
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tucaaue S. Alvares
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Yong Zhang
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ana P. Miranda Tapia
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Vivian Biancardi
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Alexis K. Katzell
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Sara M. Frangos
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Megan A. Hansen
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Alexander W. Toohey
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Carol E. Cass
- Professor Emerita, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - James D. Young
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Silvia Pagliardini
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School and New Jersey Medical School, Rutgers University, New Brunswick, NJ, United States
| | - Gregory D. Funk
- Department of Physiology, Women and Children’s Health Research Institute, Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
11
|
Saul MC, Blatti C, Yang W, Bukhari SA, Shpigler HY, Troy JM, Seward CH, Sloofman L, Chandrasekaran S, Bell AM, Stubbs L, Robinson GE, Zhao SD, Sinha S. Cross-species systems analysis of evolutionary toolkits of neurogenomic response to social challenge. GENES BRAIN AND BEHAVIOR 2018; 18:e12502. [PMID: 29968347 DOI: 10.1111/gbb.12502] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/15/2022]
Abstract
Social challenges like territorial intrusions evoke behavioral responses in widely diverging species. Recent work has showed that evolutionary "toolkits"-genes and modules with lineage-specific variations but deep conservation of function-participate in the behavioral response to social challenge. Here, we develop a multispecies computational-experimental approach to characterize such a toolkit at a systems level. Brain transcriptomic responses to social challenge was probed via RNA-seq profiling in three diverged species-honey bees, mice and three-spined stickleback fish-following a common methodology, allowing fair comparisons across species. Data were collected from multiple brain regions and multiple time points after social challenge exposure, achieving anatomical and temporal resolution substantially greater than previous work. We developed statistically rigorous analyses equipped to find homologous functional groups among these species at the levels of individual genes, functional and coexpressed gene modules, and transcription factor subnetworks. We identified six orthogroups involved in response to social challenge, including groups represented by mouse genes Npas4 and Nr4a1, as well as common modulation of systems such as transcriptional regulators, ion channels, G-protein-coupled receptors and synaptic proteins. We also identified conserved coexpression modules enriched for mitochondrial fatty acid metabolism and heat shock that constitute the shared neurogenomic response. Our analysis suggests a toolkit wherein nuclear receptors, interacting with chaperones, induce transcriptional changes in mitochondrial activity, neural cytoarchitecture and synaptic transmission after social challenge. It shows systems-level mechanisms that have been repeatedly co-opted during evolution of analogous behaviors, thus advancing the genetic toolkit concept beyond individual genes.
Collapse
Affiliation(s)
- Michael C Saul
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Charles Blatti
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Wei Yang
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Syed A Bukhari
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Interdisciplinary Informatics Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Hagai Y Shpigler
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Ecology, Evolution and Behavior, Hebrew University, Jerusalem, Israel
| | - Joseph M Troy
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Interdisciplinary Informatics Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Christopher H Seward
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Laura Sloofman
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Genetics and Genomic Sciences, Mount Sinai Health System, New York, New York
| | | | - Alison M Bell
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Interdisciplinary Informatics Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Animal Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Lisa Stubbs
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Interdisciplinary Informatics Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Gene E Robinson
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Sihai D Zhao
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Saurabh Sinha
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
12
|
Endothelial cells cope with hypoxia-induced depletion of ATP via activation of cellular purine turnover and phosphotransfer networks. Biochim Biophys Acta Mol Basis Dis 2018. [PMID: 29514048 DOI: 10.1016/j.bbadis.2018.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Intravascular ATP and adenosine have emerged as important regulators of endothelial barrier function, vascular remodeling and neovascularization at various pathological states, including hypoxia, inflammation and oxidative stress. By using human umbilical vein endothelial cells (HUVEC) and bovine vasa vasorum endothelial cells (VVEC) as representatives of macro- and microvessel phenotypes, this study was undertaken to evaluate cellular mechanisms contributing to physiological adaptation of vascular endothelium to hypoxia, with a particular emphasis on ectoenzymatic purine-converting activities and their link to intracellular ATP homeostasis and signaling pathways. Nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39), ecto-5'-nucleotidase/CD73 and ecto-adenylate kinase activities were determined by thin-layer chromatography (TLC) with 3H-labelled nucleotide substrates. Exposure of HUVEC and VVEC to 1% O2 for 4-24 h triggered rather moderate activation of ATP breakdown into adenosine via the CD39-CD73 axis. Additional TLC analysis of salvage pathways revealed the enhanced ability of hypoxic HUVEC to convert cell-incorporated [3H]adenosine into [3H]ADP/ATP. Furthermore, following a period of hypoxia, HUVEC underwent concurrent changes in intracellular signaling manifested in the depletion of putative ATP stores and targeted up-regulation of phospho-p53, p70S6K/mTOR and other tyrosine kinases. The revealed complex implication of both extrinsic and intrinsic mechanisms into a tuned hypoxia-induced control of purine homeostasis and signaling may open up further research for the development of pharmacological treatments to improve endothelial cell function under disease conditions associated with a loss of cellular ATP during oxygen deprivation.
Collapse
|
13
|
Jackson EK. Discovery and Roles of 2',3'-cAMP in Biological Systems. Handb Exp Pharmacol 2017; 238:229-252. [PMID: 26721674 DOI: 10.1007/164_2015_40] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In 2009, investigators using ultra-performance liquid chromatography-tandem mass spectrometry to measure, by selected reaction monitoring, 3',5'-cAMP in the renal venous perfusate from isolated, perfused kidneys detected a large signal at the same m/z transition (330 → 136) as 3',5'-cAMP but at a different retention time. Follow-up experiments demonstrated that this signal was due to a positional isomer of 3',5'-cAMP, namely, 2',3'-cAMP. Soon thereafter, investigative teams reported the detection of 2',3'-cAMP and other 2',3'-cNMPs (2',3'-cGMP, 2',3'-cCMP, and 2',3'-cUMP) in biological systems ranging from bacteria to plants to animals to humans. Injury appears to be the major stimulus for the release of these unique noncanonical cNMPs, which likely are formed by the breakdown of RNA. In mammalian cells in culture, in intact rat and mouse kidneys, and in mouse brains in vivo, 2',3'-cAMP is metabolized to 2'-AMP and 3'-AMP; and these AMPs are subsequently converted to adenosine. In rat and mouse kidneys and mouse brains, injury releases 2',3'-cAMP, 2'-AMP, and 3'-AMP into the extracellular compartment; and in humans, traumatic brain injury is associated with large increases in 2',3'-cAMP, 2'-AMP, 3'-AMP, and adenosine in the cerebrospinal fluid. These findings motivate the extracellular 2',3'-cAMP-adenosine pathway hypothesis: intracellular production of 2',3'-cAMP → export of 2',3'-cAMP → extracellular metabolism of 2',3'-cAMP to 2'-AMP and 3'-AMP → extracellular metabolism of 2'-AMP and 3'-AMP to adenosine. Since 2',3'-cAMP has been shown to activate mitochondrial permeability transition pores (mPTPs) leading to apoptosis and necrosis and since adenosine is generally tissue protective, the extracellular 2',3'-cAMP-adenosine pathway may be a protective mechanism [i.e., removes 2',3'-cAMP (an intracellular toxin) and forms adenosine (a tissue protectant)]. This appears to be the case in the brain where deficiency in CNPase (the enzyme that metabolizes 2',3'-cAMP to 2-AMP) leads to increased susceptibility to brain injury and neurological diseases. Surprisingly, CNPase deficiency in the kidney actually protects against acute kidney injury, perhaps by preventing the formation of 2'-AMP (which turns out to be a renal vasoconstrictor) and by augmenting the mitophagy of damaged mitochondria. With regard to 2',3'-cNMPs and their downstream metabolites, there is no doubt much more to be discovered.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
14
|
Straussberg R, Onoufriadis A, Konen O, Zouabi Y, Cohen L, Lee JYW, Hsu CK, Simpson MA, McGrath JA. Novel homozygous missense mutation in NT5C2 underlying hereditary spastic paraplegia SPG45. Am J Med Genet A 2017; 173:3109-3113. [PMID: 28884889 DOI: 10.1002/ajmg.a.38414] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/18/2017] [Accepted: 07/29/2017] [Indexed: 01/23/2023]
Abstract
SPG45 is a rare form of autosomal recessive spastic paraplegia associated with mental retardation. Detailed phenotyping and mutation analysis was undertaken in three individuals with SPG45 from a consanguineous family of Arab Muslim origin. Using whole-exome sequencing, we identified a novel homozygous missense mutation in NT5C2 (c.1379T>C; p.Leu460Pro). Our data expand the molecular basis of SPG45, adding the first missense mutation to the current database of nonsense, frameshift, and splice site mutations. NT5C2 mutations seem to have a broad clinical spectrum and should be sought in patients manifesting either as uncomplicated or complicated HSP.
Collapse
Affiliation(s)
- Rachel Straussberg
- Neurogenetic Clinic, Neurology Institute, Schneider Children's Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alexandros Onoufriadis
- St John's Institute of Dermatology, Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Osnat Konen
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Radiology Institute, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Yasmin Zouabi
- Neurogenetic Clinic, Neurology Institute, Schneider Children's Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lior Cohen
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Genetic Institute, Schneider Children's Medical Center, Petah Tikva, Israel
| | - John Y W Lee
- St John's Institute of Dermatology, Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Chao-Kai Hsu
- St John's Institute of Dermatology, Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Michael A Simpson
- Department of Medical and Molecular Genetics, Division of Genetics and Molecular Medicine, King's College London, Guy's Hospital, London, UK
| | - John A McGrath
- St John's Institute of Dermatology, Division of Genetics and Molecular Medicine, King's College London, London, UK
| |
Collapse
|
15
|
Fuentes F, Alarcón M, Badimon L, Fuentes M, Klotz KN, Vilahur G, Kachler S, Padró T, Palomo I, Fuentes E. Guanosine exerts antiplatelet and antithrombotic properties through an adenosine-related cAMP-PKA signaling. Int J Cardiol 2017; 248:294-300. [PMID: 28811090 DOI: 10.1016/j.ijcard.2017.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/09/2017] [Accepted: 08/04/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Guanosine is a natural product and an endogenous nucleoside that has shown to increase during myocardial ischemia. Platelets are critically involved in ischemic coronary events. It remains unknown, however, whether guanosine may affect platelet activation and function. We sought to investigate the potential antiplatelet and antithrombotic properties of guanosine and decipher the mechanisms behind. METHODS We firstly assessed the effects of guanosine on platelet activation/aggregation upon stimulation with several platelet agonists including adenosine diphosphate (ADP), collagen, arachidonic acid (AA), and TRAP-6. Guanosine antithrombotic potential was also evaluated both in vitro (Badimon perfusion chamber) and in vivo (murine model). In addition we assessed any potential effect on bleeding. At a mechanistic level we determined the release of thromboxane B2, intraplatelet cAMP levels, the binding affinity on platelet membrane, and the activation/phosphorylation of protein kinase A (PKA), phospholipase C (PLC) and PKC. RESULTS Guanosine markedly inhibited platelet activation/aggregation-challenged by ADP and, although to a lesser extent, also reduced platelet aggregation challenged by collagen, AA and TRAP-6. Guanosine significantly reduced thrombus formation both in vitro and in vivo without significantly affects bleeding. Guanosine antiplatelet effects were associated with the activation of the cAMP/PKA signaling pathway, and a reduction in thromboxane B2 levels and PLC and PKC phosphorylation. The platelet aggregation and binding affinity assays revealed that guanosine effects on platelets were mediated by adenosine. CONCLUSION Guanosine effectively reduces ADP-induced platelet aggregation and limits thrombotic risk. These antithrombotic properties are associated with the activation of the cAMP/PKA signaling pathway.
Collapse
Affiliation(s)
- Francisco Fuentes
- Becario Obstetricia y Ginecología, Universidad Católica del Maule, Talca, Chile
| | - Marcelo Alarcón
- Platelet Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile
| | - Lina Badimon
- Cardiovascular Science Institute - ICCC,IIB-Sant Pau, CIBERCV, Barcelona, Spain; Cardiovascular Research Chair, Universidad Autónoma Barcelona (UAB), Barcelona, Spain
| | - Manuel Fuentes
- Platelet Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile
| | - Karl-Norbert Klotz
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078 Würzburg, Germany
| | - Gemma Vilahur
- Cardiovascular Science Institute - ICCC,IIB-Sant Pau, CIBERCV, Barcelona, Spain
| | - Sonja Kachler
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078 Würzburg, Germany
| | - Teresa Padró
- Cardiovascular Science Institute - ICCC,IIB-Sant Pau, CIBERCV, Barcelona, Spain
| | - Iván Palomo
- Platelet Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile.
| | - Eduardo Fuentes
- Platelet Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile; Núcleo Científico Multidisciplinario, Universidad de Talca, Talca, Chile.
| |
Collapse
|
16
|
Thomaz DT, Dal-Cim TA, Martins WC, Cunha MP, Lanznaster D, de Bem AF, Tasca CI. Guanosine prevents nitroxidative stress and recovers mitochondrial membrane potential disruption in hippocampal slices subjected to oxygen/glucose deprivation. Purinergic Signal 2016; 12:707-718. [PMID: 27613537 DOI: 10.1007/s11302-016-9534-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/26/2016] [Indexed: 12/12/2022] Open
Abstract
Guanosine, the endogenous guanine nucleoside, prevents cellular death induced by ischemic events and is a promising neuroprotective agent. During an ischemic event, nitric oxide has been reported to either cause or prevent cell death. Our aim was to evaluate the neuroprotective effects of guanosine against oxidative damage in hippocampal slices subjected to an in vitro ischemia model, the oxygen/glucose deprivation (OGD) protocol. We also assessed the participation of nitric oxide synthase (NOS) enzymes activity on the neuroprotection promoted by guanosine. Here, we showed that guanosine prevented the increase in ROS, nitric oxide, and peroxynitrite production induced by OGD. Moreover, guanosine prevented the loss of mitochondrial membrane potential in hippocampal slices subjected to OGD. Guanosine did not present an antioxidant effect per se. The protective effects of guanosine were mimicked by inhibition of neuronal NOS, but not of inducible NOS. The neuroprotective effect of guanosine may involve activation of cellular mechanisms that prevent the increase in nitric oxide production, possibly via neuronal NOS.
Collapse
Affiliation(s)
- Daniel T Thomaz
- Programa de Pós-Graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Tharine A Dal-Cim
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Wagner C Martins
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Maurício Peña Cunha
- Programa de Pós-Graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Débora Lanznaster
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Andreza F de Bem
- Departamento de Bioquímica, CCB, UFSC, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brazil
- Programa de Pós-Graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Carla I Tasca
- Departamento de Bioquímica, CCB, UFSC, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brazil.
- Programa de Pós-Graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
17
|
Bettio LEB, Gil-Mohapel J, Rodrigues ALS. Guanosine and its role in neuropathologies. Purinergic Signal 2016; 12:411-26. [PMID: 27002712 PMCID: PMC5023624 DOI: 10.1007/s11302-016-9509-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/08/2016] [Indexed: 02/08/2023] Open
Abstract
Guanosine is a purine nucleoside thought to have neuroprotective properties. It is released in the brain under physiological conditions and even more during pathological events, reducing neuroinflammation, oxidative stress, and excitotoxicity, as well as exerting trophic effects in neuronal and glial cells. In agreement, guanosine was shown to be protective in several in vitro and/or in vivo experimental models of central nervous system (CNS) diseases including ischemic stroke, Alzheimer's disease, Parkinson's disease, spinal cord injury, nociception, and depression. The mechanisms underlying the neurobiological properties of guanosine seem to involve the activation of several intracellular signaling pathways and a close interaction with the adenosinergic system, with a consequent stimulation of neuroprotective and regenerative processes in the CNS. Within this context, the present review will provide an overview of the current literature on the effects of guanosine in the CNS. The elucidation of the complex signaling events underlying the biochemical and cellular effects of this nucleoside may further establish guanosine as a potential therapeutic target for the treatment of several neuropathologies.
Collapse
Affiliation(s)
- Luis E B Bettio
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil.
| |
Collapse
|
18
|
Kawabori M, Yenari MA. Inflammatory responses in brain ischemia. Curr Med Chem 2016; 22:1258-77. [PMID: 25666795 DOI: 10.2174/0929867322666150209154036] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/02/2014] [Accepted: 02/02/2015] [Indexed: 12/20/2022]
Abstract
Brain infarction causes tissue death by ischemia due to occlusion of the cerebral vessels and recent work has shown that post stroke inflammation contributes significantly to the development of ischemic pathology. Because secondary damage by brain inflammation may have a longer therapeutic time window compared to the rescue of primary damage following arterial occlusion, controlling inflammation would be an obvious therapeutic target. A substantial amount of experimentall progress in this area has been made in recent years. However, it is difficult to elucidate the precise mechanisms of the inflammatory responses following ischemic stroke because inflammation is a complex series of interactions between inflammatory cells and molecules, all of which could be either detrimental or beneficial. We review recent advances in neuroinflammation and the modulation of inflammatory signaling pathways in brain ischemia. Potential targets for treatment of ischemic stroke will also be covered. The roles of the immune system and brain damage versus repair will help to clarify how immune modulation may treat stroke.
Collapse
Affiliation(s)
| | - Midori A Yenari
- Dept. of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA.
| |
Collapse
|
19
|
Beart PM. Synaptic signalling and its interface with neuropathologies: snapshots from the past, present and future. J Neurochem 2016; 139 Suppl 2:76-90. [PMID: 27144305 DOI: 10.1111/jnc.13598] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/09/2016] [Accepted: 02/26/2016] [Indexed: 11/30/2022]
Abstract
This 'Past to Future' Review as part of the 60th anniversary year of the Journal of Neurochemistry focuses on synaptic transmission and associated signalling, and seeks to identify seminal progress in neurochemistry over the last 10 years which has advanced our understanding of neuronal communication in brain. The approach adopted analyses neurotransmitters on a case by case basis (i.e. amino acids, monoamines, acetylcholine, neuropeptides, ATP/purines and gasotransmitters) to highlight novel findings that have changed the way we view each type of transmitter, to explore commonalities and interactions, and to note how new insights have changed the way we view the biology of degenerative, psychiatric and behavioural conditions. Across all transmitter systems there was remarkable growth in the identification of targets likely to provide therapeutic benefit and which undoubtedly was driven by the elucidation of circuit function and new vistas of synaptic signalling. There has been an increasing trend to relate signalling to disease, notably for Alzheimer's and Parkinson's disease and related conditions, and which has occurred for each transmitter family. Forebrain circuitry and tonic excitatory control have been the centre of great attention yielding novel findings that will impact upon cognitive, emotional and addictive behaviours. Other impressive insights focus on gasotransmitters integrating activity as volume transmitters. Exciting developments in how serotonin, cholinergic, l-glutamate, galanin and adenosine receptors and their associated signalling can be beneficially targeted should underpin the development of new therapies. Clearly integrated, multifaceted neurochemistry has changed the way we view synaptic signalling and its relevance to pathobiology. Highlighted are important advances in synaptic signalling over the last decade in the Journal of Neurochemistry. Across all transmitter systems elucidation of circuit function, and notably molecular insights, have underpinned remarkable growth in the identification of targets likely to provide therapeutic benefit in neuropathologies. Another commonality was wide interest in forebrain circuitry and its tonic excitatory control. Increasingly observations relate to signalling in disease and behavioural conditions. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Philip M Beart
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
20
|
Stockwell J, Chen Z, Niazi M, Nosib S, Cayabyab FS. Protein phosphatase role in adenosine A1 receptor-induced AMPA receptor trafficking and rat hippocampal neuronal damage in hypoxia/reperfusion injury. Neuropharmacology 2015; 102:254-65. [PMID: 26626486 DOI: 10.1016/j.neuropharm.2015.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/19/2015] [Accepted: 11/21/2015] [Indexed: 12/19/2022]
Abstract
Adenosine signaling via A1 receptor (A1R) and A2A receptor (A2AR) has shown promise in revealing potential targets for neuroprotection in cerebral ischemia. We recently showed a novel mechanism by which A1R activation with N(6)-cyclopentyl adenosine (CPA) induced GluA1 and GluA2 AMPA receptor (AMPAR) endocytosis and adenosine-induced persistent synaptic depression (APSD) in rat hippocampus. This study further investigates the mechanism of A1R-mediated AMPAR internalization and hippocampal slice neuronal damage through activation of protein phosphatase 1 (PP1), 2A (PP2A), and 2B (PP2B) using electrophysiological, biochemical and imaging techniques. Following prolonged A1R activation, GluA2 internalization was selectively blocked by PP2A inhibitors (okadaic acid and fostriecin), whereas inhibitors of PP2A, PP1 (tautomycetin), and PP2B (FK506) all prevented GluA1 internalization. Additionally, GluA1 phosphorylation at Ser831 and Ser845 was reduced after prolonged A1R activation in hippocampal slices. PP2A inhibitors nullified A1R-mediated downregulation of pSer845-GluA1, while PP1 and PP2B inhibitors prevented pSer831-GluA1 downregulation. Each protein phosphatase inhibitor also blunted CPA-induced synaptic depression and APSD. We then tested whether A1R-mediated changes in AMPAR trafficking and APSD contribute to hypoxia-induced neuronal injury. Hypoxia (20 min) induced A1R-mediated internalization of both AMPAR subunits, and subsequent normoxic reperfusion (45 min) increased GluA1 but persistently reduced GluA2 surface expression. Neuronal damage after hypoxia-reperfusion injury was significantly blunted by pre-incubation with the above protein phosphatase inhibitors. Together, these data suggest that A1R-mediated protein phosphatase activation causes persistent synaptic depression by downregulating GluA2-containing AMPARs; this previously undefined role of A1R stimulation in hippocampal neuronal damage represents a novel therapeutic target in cerebral ischemic damage.
Collapse
Affiliation(s)
- Jocelyn Stockwell
- Department of Surgery, Neuroscience Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Zhicheng Chen
- Department of Surgery, Neuroscience Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Mina Niazi
- Department of Surgery, Neuroscience Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Siddarth Nosib
- Department of Surgery, Neuroscience Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Francisco S Cayabyab
- Department of Surgery, Neuroscience Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
21
|
Region-specific metabolic alterations in the brain of the APP/PS1 transgenic mice of Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2395-402. [DOI: 10.1016/j.bbadis.2014.09.014] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/22/2014] [Accepted: 09/28/2014] [Indexed: 11/21/2022]
|
22
|
Zhao LR, Piao CS, Murikinati SR, Gonzalez-Toledo ME. The role of stem cell factor and granulocyte-colony stimulating factor in treatment of stroke. ACTA ACUST UNITED AC 2014; 8:2-12. [PMID: 23173646 DOI: 10.2174/1574889811308010002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Revised: 11/02/2012] [Accepted: 11/02/2012] [Indexed: 11/22/2022]
Abstract
Stroke is a serious cerebrovascular disease that causes high mortality and persistent disability in adults worldwide. Stroke is also an enormous public health problem and a heavy public financial burden in the United States. Treatment for stroke is very limited. Thrombolytic therapy by tissue plasminogen activator (tPA) is the only approved treatment for acute stroke, and no effective treatment is available for chronic stroke. Developing new therapeutic strategies, therefore, is a critical need for stroke treatment. This article summarizes the discovery of new routes of treatment for acute and chronic stroke using two hematopoietic growth factors, stem cell factor (SCF) and granulocyte-colony stimulating factor (G-CSF). In a study of acute stroke, SCF and G-CSF alone or in combination displays neuroprotective effects in an animal model of stroke. SCF appears to be the optimal treatment for acute stroke as the functional outcome is superior to G-CSF alone or in combination (SCF+G-CSF); however, SCF+G-CSF does show better functional recovery than G-CSF. In a chronic stroke study, the therapeutic effects of SCF and G-CSF alone or in combination appear differently as compared with their effects on the acute stroke. SCF+G-CSF induces stable and long-lasting functional improvement; SCF alone also improves functional outcome but its effectiveness is less than SCF+G-CSF, whereas G-CSF shows no therapeutic effects. Although the mechanism by which SCF+G-CSF repairs the brain in chronic stroke remains poorly understood, our recent findings suggest that the SCF+G-CSF-induced functional improvement in chronic stroke is associated with a contribution to increasing angiogenesis and neurogenesis through bone marrow-derived cells and the direct effects on stimulating neurons to form new neuronal networks. These findings would assist in developing new treatment for stroke. The article presents some promising patents on role of stem cell factor and granulocyte-colony stimulating factor in treatment of stroke.
Collapse
Affiliation(s)
- Li R Zhao
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, 1501 Kings Highway, Shreveport, Louisiana 71130, USA.
| | | | | | | |
Collapse
|
23
|
Antonioli L, Csóka B, Fornai M, Colucci R, Kókai E, Blandizzi C, Haskó G. Adenosine and inflammation: what's new on the horizon? Drug Discov Today 2014; 19:1051-68. [DOI: 10.1016/j.drudis.2014.02.010] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 02/06/2014] [Accepted: 02/25/2014] [Indexed: 12/18/2022]
|
24
|
Senyavina NV, Khaustova SA, Grebennik TK, Pavlovich SV. Analysis of purine metabolites in maternal serum for evaluating the risk of gestosis. Bull Exp Biol Med 2014; 155:682-4. [PMID: 24288739 DOI: 10.1007/s10517-013-2225-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Metabolome analysis of the serum from pregnant patients aimed at detection of low-molecular-weight biomarkers of gestation process disorders indicated a relationship between the metabolic profile of maternal serum and risk of gestosis. In women with pre-eclampsia or preterm delivery, analysis of serum purine metabolites revealed changes in the metabolite concentrations, associated with pregnancy complications.
Collapse
Affiliation(s)
- N V Senyavina
- BioClinicum Center; V. I. Kulakov Center of Obstetrics, Gynecology, and Perinatology, the Ministry of Health and Social Development of the Russian Federation, Moscow, Russia.
| | | | | | | |
Collapse
|
25
|
Abstract
In cultured renal cells and isolated perfused kidneys, extracellular guanosine augments extracellular adenosine and inosine (the major renal metabolite of adenosine) levels by altering the extracellular disposition of these purines. The present study addressed whether this "guanosine-adenosine mechanism" exists in vivo. In rats (n = 15), intravenous infusions of adenosine (1 µmol/kg per minute) decreased mean arterial blood pressure (MABP) from 114 ± 4 to 83 ± 5 mm Hg, heart rate (HR) from 368 ± 11 to 323 ± 9 beats/min), and renal blood flow (RBF) from 6.2 ± 0.5 to 5.3 ± 0.6 ml/min). In rats (n = 15) pretreated with intravenous guanosine (10 µmol/kg per minute), intravenous adenosine (1 µmol/kg per minute) decreased MABP (from 109 ± 4 to 58 ± 5 mm Hg), HR (from 401 ± 10 to 264 ± 20 beats/min), and RBF (from 6.2 ± 0.7 to 1.7 ± 0.3). Two-factor analysis of variance (2F-ANOVA) revealed a significant interaction (P < 0.0001) between guanosine and adenosine for MABP, HR, and RBF. In control rats, the urinary excretion rate of endogenous inosine was 211 ± 103 ng/30 minutes (n = 9); however, in rats treated with intravenous guanosine (10 µmol/kg per minute), the excretion rate of inosine was 1995 ± 300 ng/30 minutes (n = 12; P < 0.0001 versus controls). Because adenosine inhibits inflammatory cytokine production, we also examined the effects of intravenous guanosine on endotoxemia-induced increases in tumor necrosis factor-α (TNF-α). In control rats (n = 7), lipopolysaccharide (LPS; Escherichia coli 026:B6 endotoxin; 30 mg/kg) increased plasma TNF-α from 164 ± 56 to 4082 ± 730 pg/ml, whereas in rats pretreated with intravenous guanosine (10 µmol/kg per minute; n = 6), LPS increased plasma TNF-α from 121 ± 45 to 1821 ± 413 pg/ml (2F-ANOVA interaction effect, P = 0.0022). We conclude that the guanosine-adenosine mechanism exists in vivo and that guanosine may be a useful therapeutic for reducing inflammation.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
26
|
Abstract
In cell culture, extracellular guanosine increases extracellular adenosine by attenuating the disposition of extracellular adenosine (American Journal of Physiology – Cell Physiology 304: C406–C421, 2013). The goal of this investigation was to determine whether this “guanosine–adenosine mechanism” is operative in an intact organ. Twenty‐seven isolated, perfused mouse kidneys were subjected to metabolic poisons (iodoacetate plus 2,4‐dinitrophenol) to cause energy depletion and thereby stimulate renal adenosine production. Adenosine levels in the renal venous perfusate increased from a baseline of 36 ± 8 to 499 ± 96, 258 ± 50, and 71 ± 13 nmol/L at 15, 30, and 60 min, respectively, after administering metabolic poisons (% of basal; 1366 ± 229, 715 ± 128, and 206 ± 33, respectively). Changes in renal venous levels of guanosine closely mirrored the time course of changes in adenosine: baseline of 15 ± 2 to 157 ± 13, 121 ± 8, and 50 ± 5 nmol/L at 15, 30, and 60 min, respectively (% of basal; 1132 ± 104, 871 ± 59, and 400 ± 51, respectively). Freeze‐clamp experiments in 12 kidneys confirmed that metabolic poisons increased kidney tissue levels of adenosine and guanosine. In eight additional kidneys, we examined the ability of guanosine to reduce the renal clearance of exogenous adenosine; and these experiments revealed that guanosine significantly decreased the renal extraction of adenosine. Because guanosine is metabolized by purine nucleoside phosphorylase (PNPase), in another set of 16 kidneys we examined the effects of 8‐aminoguanine (PNPase inhibitor) on renal venous levels of adenosine and inosine (adenosine metabolite). Kidneys treated with 8‐aminoguanine showed a more robust increase in both adenosine and inosine in response to metabolic poisons. We conclude that in the intact kidney, guanosine regulates adenosine levels. In cell culture, extracellular guanosine increases extracellular adenosine by attenuating the disposition of extracellular adenosine (American Journal of Physiology – Cell Physiology 304: C406–C421, 2013). The goal of this study was to determine whether the “guanosine–adenosine mechanism” is operative in an intact organ. In isolated, perfused mouse kidneys, inhibition of energy production induced changes in renal venous levels of guanosine that closely mirrored the time course of changes in adenosine, and freeze‐clamp experiments confirmed that metabolic poisons similarly increased kidney tissue levels of adenosine and guanosine. Moreover, exogenous guanosine significantly decreased the renal extraction of exogenous adenosine, and inhibition of purine nucleoside phosphorylase (metabolizes guanosine) augmented the effects of energy depletion on renal levels of both guanosine and adenosine. We conclude that in the intact kidney, guanosine regulates adenosine levels.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dongmei Cheng
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Delbert G Gillespie
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
27
|
Thauerer B, Zur Nedden S, Baier-Bitterlich G. Protein Kinase C-Related Kinase (PKN/PRK). Potential Key-Role for PKN1 in Protection of Hypoxic Neurons. Curr Neuropharmacol 2014; 12:213-8. [PMID: 24851086 PMCID: PMC4023452 DOI: 10.2174/1570159x11666131225000518] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/20/2013] [Accepted: 12/10/2013] [Indexed: 12/13/2022] Open
Abstract
Serine/threonine protein kinase C-related kinase (PKN/PRK) is a family of three isoenzymes (PKN1, PKN2,
PKN3), which are widely distributed in eukaryotic organisms and share the same overall domain structure. The Nterminal
region encompasses a conserved repeated domain, termed HR1a-c as well as a HR2/C2 domain. The
serine/threonine kinase domain is found in the C-terminal region of the protein and shows high sequence homology to
other members of the PKC superfamily.
In neurons, PKN1 is the most abundant isoform and has been implicated in a variety of functions including cytoskeletal
organization and neuronal differentiation and its deregulation may contribute to neuropathological processes such as
amyotrophic lateral sclerosis and Alzheimer’s disease. We have recently identified a candidate role of PKN1 in the
regulation of neuroprotective processes during hypoxic stress. Our key findings were that: 1) the activity of PKN1 was
significantly increased by hypoxia (1% O2) and neurotrophins (nerve growth factor and purine nucleosides); 2) Neuronal
cells, deficient of PKN1 showed a decrease of cell viability and neurite formation along with a disturbance of the F-actinassociated
cytoskeleton; 3) Purine nucleoside-mediated neuroprotection during hypoxia was severely hampered in PKN1
deficient neuronal cells, altogether suggesting a potentially critical role of PKN1 in neuroprotective processes.
This review gives an up-to-date overview of the PKN family with a special focus on the neuroprotective role of PKN1 in
hypoxia.
Collapse
Affiliation(s)
- Bettina Thauerer
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Stephanie Zur Nedden
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Gabriele Baier-Bitterlich
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| |
Collapse
|
28
|
Novarino G, Fenstermaker AG, Zaki MS, Hofree M, Silhavy JL, Heiberg AD, Abdellateef M, Rosti B, Scott E, Mansour L, Masri A, Kayserili H, Al-Aama JY, Abdel-Salam GMH, Karminejad A, Kara M, Kara B, Bozorgmehri B, Ben-Omran T, Mojahedi F, El Din Mahmoud IG, Bouslam N, Bouhouche A, Benomar A, Hanein S, Raymond L, Forlani S, Mascaro M, Selim L, Shehata N, Al-Allawi N, Bindu P, Azam M, Gunel M, Caglayan A, Bilguvar K, Tolun A, Issa MY, Schroth J, Spencer EG, Rosti RO, Akizu N, Vaux KK, Johansen A, Koh AA, Megahed H, Durr A, Brice A, Stevanin G, Gabriel SB, Ideker T, Gleeson JG. Exome sequencing links corticospinal motor neuron disease to common neurodegenerative disorders. Science 2014; 343:506-511. [PMID: 24482476 PMCID: PMC4157572 DOI: 10.1126/science.1247363] [Citation(s) in RCA: 412] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hereditary spastic paraplegias (HSPs) are neurodegenerative motor neuron diseases characterized by progressive age-dependent loss of corticospinal motor tract function. Although the genetic basis is partly understood, only a fraction of cases can receive a genetic diagnosis, and a global view of HSP is lacking. By using whole-exome sequencing in combination with network analysis, we identified 18 previously unknown putative HSP genes and validated nearly all of these genes functionally or genetically. The pathways highlighted by these mutations link HSP to cellular transport, nucleotide metabolism, and synapse and axon development. Network analysis revealed a host of further candidate genes, of which three were mutated in our cohort. Our analysis links HSP to other neurodegenerative disorders and can facilitate gene discovery and mechanistic understanding of disease.
Collapse
Affiliation(s)
- Gaia Novarino
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ali G. Fenstermaker
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maha S. Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo 12311, Egypt
| | - Matan Hofree
- Department of Computer Science and Engineering and Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jennifer L. Silhavy
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrew D. Heiberg
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mostafa Abdellateef
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Basak Rosti
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric Scott
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lobna Mansour
- Department of Pediatric Neurology, Neurometabolic Unit, Cairo University Children’s Hospital, Cairo 406, Egypt
| | - Amira Masri
- Division of Child Neurology, Department of Pediatrics, University of Jordan, Amman 11942, Jordan
| | - Hulya Kayserili
- Istanbul University, Istanbul Medical Faculty, Medical Genetics Department, 34093 Istanbul, Turkey
| | - Jumana Y. Al-Aama
- Department of Genetic Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Ghada M. H. Abdel-Salam
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo 12311, Egypt
| | | | - Majdi Kara
- Department of Pediatrics, Tripoli Children’s Hospital, Tripoli, Libya
| | - Bulent Kara
- Kocaeli University, Medical Faculty, Department of Pediatric Neurology, 41380 Umuttepe, Kocaeli, Turkey
| | - Bita Bozorgmehri
- Kariminejad-Najmabadi Pathology and Genetics Center, Tehran, Iran
| | - Tawfeg Ben-Omran
- Clinical and Metabolic Genetics Division, Department of Pediatrics, Hamad Medical Corporation, Doha 3050, Qatar
| | - Faezeh Mojahedi
- Mashhad Medical Genetic Counseling Center, 91767 Mashhad, Iran
| | - Iman Gamal El Din Mahmoud
- Department of Pediatric Neurology, Neurometabolic Unit, Cairo University Children’s Hospital, Cairo 406, Egypt
| | - Naima Bouslam
- Université Mohammed V Souissi, Equipe de Recherchéde Maladies Neurodégéneratives (ERMN) and Centre de Recherche en Épidémiologie Clinique et Essais Thérapeutiques (CRECET), 6402 Rabat, Morocco
| | - Ahmed Bouhouche
- Université Mohammed V Souissi, Equipe de Recherchéde Maladies Neurodégéneratives (ERMN) and Centre de Recherche en Épidémiologie Clinique et Essais Thérapeutiques (CRECET), 6402 Rabat, Morocco
| | - Ali Benomar
- Université Mohammed V Souissi, Equipe de Recherchéde Maladies Neurodégéneratives (ERMN) and Centre de Recherche en Épidémiologie Clinique et Essais Thérapeutiques (CRECET), 6402 Rabat, Morocco
| | - Sylvain Hanein
- Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière, INSERM U1127, CNRS UMR7225; UPMC Univ Paris VI UMR_S975, 75013 Paris, France
| | - Laure Raymond
- Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière, INSERM U1127, CNRS UMR7225; UPMC Univ Paris VI UMR_S975, 75013 Paris, France
| | - Sylvie Forlani
- Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière, INSERM U1127, CNRS UMR7225; UPMC Univ Paris VI UMR_S975, 75013 Paris, France
| | - Massimo Mascaro
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Laila Selim
- Department of Pediatric Neurology, Neurometabolic Unit, Cairo University Children’s Hospital, Cairo 406, Egypt
| | - Nabil Shehata
- Department of Pediatrics and Neonatology, Saudi German Hospital, Post Office Box 84348, Riyadh, Kingdom of Saudi Arabia
| | - Nasir Al-Allawi
- Department of Pathology, School of Medicine, University of Dohuk, Dohuk, Iraq
| | - P.S. Bindu
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Matloob Azam
- Department of Pediatrics and Child Neurology, Wah Medical College, Wah Cantt, Pakistan
| | - Murat Gunel
- Department of Genetics and Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ahmet Caglayan
- Department of Genetics and Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kaya Bilguvar
- Department of Genetics and Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Aslihan Tolun
- Department of Molecular Biology and Genetics, Bogazici University, 34342 Istanbul, Turkey
| | - Mahmoud Y. Issa
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo 12311, Egypt
| | - Jana Schroth
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emily G. Spencer
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rasim O. Rosti
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Naiara Akizu
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Keith K. Vaux
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anide Johansen
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alice A. Koh
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hisham Megahed
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo 12311, Egypt
| | - Alexandra Durr
- Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière, INSERM U1127, CNRS UMR7225; UPMC Univ Paris VI UMR_S975, 75013 Paris, France
- Assistance Publique–Hôpitaux de Paris, Fédération de Génétique, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Alexis Brice
- Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière, INSERM U1127, CNRS UMR7225; UPMC Univ Paris VI UMR_S975, 75013 Paris, France
- Assistance Publique–Hôpitaux de Paris, Fédération de Génétique, Pitié-Salpêtrière Hospital, 75013 Paris, France
- Institut du Cerveau et de la Moelle Épinière, 75013 Paris, France
| | - Giovanni Stevanin
- Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière, INSERM U1127, CNRS UMR7225; UPMC Univ Paris VI UMR_S975, 75013 Paris, France
- Assistance Publique–Hôpitaux de Paris, Fédération de Génétique, Pitié-Salpêtrière Hospital, 75013 Paris, France
- Institut du Cerveau et de la Moelle Épinière, 75013 Paris, France
- Laboratoire de Neurogénétique, Ecole Pratique des Hautes Etudes, Institut du Cerveau et de la Moelle Épinière, 75013 Paris, France
| | - Stacy B. Gabriel
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Trey Ideker
- Department of Computer Science and Engineering and Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joseph G. Gleeson
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
29
|
Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev 2013; 66:102-92. [PMID: 24335194 DOI: 10.1124/pr.113.008029] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purinergic signaling plays important roles in control of vascular tone and remodeling. There is dual control of vascular tone by ATP released as a cotransmitter with noradrenaline from perivascular sympathetic nerves to cause vasoconstriction via P2X1 receptors, whereas ATP released from endothelial cells in response to changes in blood flow (producing shear stress) or hypoxia acts on P2X and P2Y receptors on endothelial cells to produce nitric oxide and endothelium-derived hyperpolarizing factor, which dilates vessels. ATP is also released from sensory-motor nerves during antidromic reflex activity to produce relaxation of some blood vessels. In this review, we stress the differences in neural and endothelial factors in purinergic control of different blood vessels. The long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides in promoting migration and proliferation of both vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis and vessel remodeling during restenosis after angioplasty are described. The pathophysiology of blood vessels and therapeutic potential of purinergic agents in diseases, including hypertension, atherosclerosis, ischemia, thrombosis and stroke, diabetes, and migraine, is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; and Department of Pharmacology, The University of Melbourne, Australia.
| | | |
Collapse
|
30
|
Jackson EK, Gillespie DG. Regulation of Cell Proliferation by the Guanosine-Adenosine Mechanism: Role of Adenosine Receptors. Physiol Rep 2013; 1:e00024. [PMID: 23956837 PMCID: PMC3743120 DOI: 10.1002/phy2.24] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A recent study (American Journal of Physiology – Cell Physiology 304:C406–C421, 2013) suggests that extracellular guanosine increases extracellular adenosine by modifying the disposition of extracellular adenosine (“guanosine–adenosine mechanism”) and that the guanosine–adenosine mechanism is not mediated by classical adenosine transport systems (SLC28 and SLC29 families) nor by classical adenosine-metabolizing enzymes. The present investigation had two aims (1) to test the hypothesis that the “guanosine–adenosine mechanism” affects cell proliferation; and (2) to determine whether the transporters SLC19A1, SLC19A2, SLC19A3, or SLC22A2 (known to carrier guanosine analogs) might be responsible for the guanosine–adenosine mechanism. In the absence of added adenosine, guanosine had little effect on the proliferation of coronary artery vascular smooth muscle cells (vascular conduit cells) or preglomerular vascular smooth muscle cells (vascular resistance cells). However, in the presence of added adenosine (3 or 10 μmol/L), guanosine (10–100 μmol/L) decreased proliferation of both cell types, thus resulting in a highly significant (P < 0.000001) interaction between guanosine and adenosine on cell proliferation. The guanosine–adenosine interaction on cell proliferation was abolished by 1,3-dipropyl-8-(p-sulfophenyl)xanthine (adenosine receptor antagonist). Guanosine (30 μmol/L) increased extracellular levels of adenosine when adenosine (3 μmol/L) was added to the medium. This effect was not reproduced by high concentrations of methotrexate (100 μmol/L), thiamine (1000 μmol/L), chloroquine (1000 μmol/L), or acyclovir (10,000 μmol/L), archetypal substrates for SLC19A1, SLC19A2, SLC19A3, and SLC22A2, respectively; and guanosine still increased adenosine levels in the presence of these compounds. In conclusion, the guanosine–adenosine mechanism affects cell proliferation and is not mediated by SLC19A1, SLC19A2, SLC19A3, or SLC22A2.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15219
| | | |
Collapse
|
31
|
He X, Sandhu HK, Yang Y, Hua F, Belser N, Kim DH, Xia Y. Neuroprotection against hypoxia/ischemia: δ-opioid receptor-mediated cellular/molecular events. Cell Mol Life Sci 2013; 70:2291-303. [PMID: 23014992 PMCID: PMC11113157 DOI: 10.1007/s00018-012-1167-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 09/08/2012] [Accepted: 09/10/2012] [Indexed: 12/24/2022]
Abstract
Hypoxic/ischemic injury remains the most dreaded cause of neurological disability and mortality. Despite the humbling experiences due to lack of promising therapy, our understanding of the complex cascades underlying the neuronal insult has led to advances in basic science research. One of the most noteworthy has been the effect of opioid receptors, especially the delta-opioid receptor (DOR), on hypoxic/ischemic neurons. Our recent studies, and those of others worldwide, present strong evidence that sheds light on DOR-mediated neuroprotection in the brain, especially in the cortex. The mechanisms of DOR neuroprotection are broadly categorized as: (1) stabilization of the ionic homeostasis, (2) inhibition of excitatory transmitter release, (3) attenuation of disrupted neuronal transmission, (4) increase in antioxidant capacity, (5) regulation of intracellular pathways-inhibition of apoptotic signals and activation of pro-survival signaling, (6) regulation of specific gene and protein expression, and (7) up-regulation of endogenous opioid release and/or DOR expression. Depending upon the severity and duration of hypoxic/ischemic insult, the release of endogenous opioids and DOR expression are regulated in response to the stress, and DOR signaling acts at multiple levels to confer neuronal tolerance to harmful insult. The phenomenon of DOR neuroprotection offers a potential clue for a promising target that may have significant clinical implications in our quest for neurotherapeutics.
Collapse
Affiliation(s)
- Xiaozhou He
- The Third Clinical College of Suzhou University, Changzhou, Jiangsu China
| | - Harleen K. Sandhu
- The Vivian L Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, 77030 TX USA
| | - Yilin Yang
- The Third Clinical College of Suzhou University, Changzhou, Jiangsu China
| | - Fei Hua
- The Third Clinical College of Suzhou University, Changzhou, Jiangsu China
| | - Nathalee Belser
- The Vivian L Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, 77030 TX USA
| | - Dong H. Kim
- The Vivian L Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, 77030 TX USA
| | - Ying Xia
- The Vivian L Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, 77030 TX USA
| |
Collapse
|
32
|
Dal-Cim T, Ludka FK, Martins WC, Reginato C, Parada E, Egea J, López MG, Tasca CI. Guanosine controls inflammatory pathways to afford neuroprotection of hippocampal slices under oxygen and glucose deprivation conditions. J Neurochem 2013; 126:437-50. [DOI: 10.1111/jnc.12324] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/02/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Tharine Dal-Cim
- Departamento de Bioquímica; Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC Brazil
| | - Fabiana K. Ludka
- Departamento de Bioquímica; Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC Brazil
- Curso de Farmácia; Universidade do Contestado; Canoinhas SC Brazil
| | - Wagner C. Martins
- Departamento de Bioquímica; Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC Brazil
| | - Charlise Reginato
- Departamento de Bioquímica; Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC Brazil
| | - Esther Parada
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
| | - Javier Egea
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
| | - Manuela G. López
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
| | - Carla I. Tasca
- Departamento de Bioquímica; Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC Brazil
| |
Collapse
|
33
|
Albrecht P, Henke N, Tien MLT, Issberner A, Bouchachia I, Maher P, Lewerenz J, Methner A. Extracellular cyclic GMP and its derivatives GMP and guanosine protect from oxidative glutamate toxicity. Neurochem Int 2013; 62:610-9. [PMID: 23357478 DOI: 10.1016/j.neuint.2013.01.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 01/13/2013] [Accepted: 01/18/2013] [Indexed: 11/15/2022]
Abstract
Cell death in response to oxidative stress plays a role in a variety of neurodegenerative diseases and can be studied in detail in the neuronal cell line HT22, where extracellular glutamate causes glutathione depletion by inhibition of the glutamate/cystine antiporter system xc(-), elevation of reactive oxygen species and eventually programmed cell death caused by cytotoxic calcium influx. Using this paradigm, we screened 54 putative extracellular peptide or small molecule ligands for effects on cell death and identified extracellular cyclic guanosine monophosphate (cGMP) as a protective substance. Extracellular cGMP was protective, whereas the cell-permeable cGMP analog 8-pCPT-cGMP or the inhibition of cGMP degradation by phosphodiesterases was toxic. Interestingly, metabolites GMP and guanosine were even more protective than cGMP and the inhibition of the conversion of GMP to guanosine attenuated its effect, suggesting that GMP offers protection through its conversion to guanosine. Guanosine increased system xc(-) activity and cellular glutathione levels in the presence of glutamate, which can be explained by transcriptional upregulation of xCT, the functional subunit of system xc(-). However, guanosine also provided protection when added late in the cell death cascade and significantly reduced the number of calcium peaking cells, which was most likely not mediated by transcriptional mechanisms. We observed no changes in the classical protective pathways such as phosphorylation of Akt, ERK1/2 or induction of Nrf2 or ATF4. We conclude that extracellular guanosine protects against endogenous oxidative stress by two probably independent mechanisms involving system xc(-) induction and inhibition of cytotoxic calcium influx.
Collapse
Affiliation(s)
- Philipp Albrecht
- Department of Neurology, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Jackson EK, Cheng D, Jackson TC, Verrier JD, Gillespie DG. Extracellular guanosine regulates extracellular adenosine levels. Am J Physiol Cell Physiol 2012; 304:C406-21. [PMID: 23242185 DOI: 10.1152/ajpcell.00212.2012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The aim of this investigation was to test the hypothesis that extracellular guanosine regulates extracellular adenosine levels. Rat preglomerular vascular smooth muscle cells were incubated with adenosine, guanosine, or both. Guanosine (30 μmol/l) per se had little effect on extracellular adenosine levels. Extracellular adenosine levels 1 h after addition of adenosine (3 μmol/l) were 0.125 ± 0.020 μmol/l, indicating rapid disposition of extracellular adenosine. Extracellular adenosine levels 1 h after addition of adenosine (3 μmol/l) plus guanosine (30 μmol/l) were 1.173 ± 0.061 μmol/l, indicating slow disposition of extracellular adenosine. Cell injury increased extracellular levels of endogenous adenosine and guanosine, and the effects of cell injury on endogenous extracellular adenosine were modulated by altering the levels of endogenous extracellular guanosine with exogenous purine nucleoside phosphorylase (converts guanosine to guanine) or 8-aminoguanosine (inhibits purine nucleoside phosphorylase). Extracellular guanosine also slowed the disposition of extracellular adenosine in rat preglomerular vascular endothelial cells, mesangial cells, cardiac fibroblasts, and kidney epithelial cells and in human aortic and coronary artery vascular smooth muscle cells and coronary artery endothelial cells. The effects of guanosine on adenosine levels were not mimicked or attenuated by 5-iodotubericidin (adenosine kinase inhibitor), erythro-9-(2-hydroxy-3-nonyl)-adenine (adenosine deaminase inhibitor), 5-aminoimidazole-4-carboxamide (guanine deaminase inhibitor), aristeromycin (S-adenosylhomocysteine hydrolase inhibitor), low sodium (inhibits concentrative nucleoside transporters), S-(4-nitrobenzyl)-6-thioinosine [inhibits equilibrative nucleoside transporter (ENT) type 1], zidovudine (inhibits ENT type 2), or acadesine (known modulator of adenosine levels). Guanosine also increases extracellular inosine, uridine, thymidine, and cytidine, yet decreases extracellular uric acid. In conclusion, extracellular guanosine regulates extracellular adenosine levels.
Collapse
Affiliation(s)
- Edwin K Jackson
- Dept. of Pharmacology and Chemical Biology, 100 Technology Drive, Rm. 514, Univ. of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| | | | | | | | | |
Collapse
|
35
|
Dal-Cim T, Molz S, Egea J, Parada E, Romero A, Budni J, Martín de Saavedra MD, Barrio LD, Tasca CI, López MG. Guanosine protects human neuroblastoma SH-SY5Y cells against mitochondrial oxidative stress by inducing heme oxigenase-1 via PI3K/Akt/GSK-3β pathway. Neurochem Int 2012; 61:397-404. [DOI: 10.1016/j.neuint.2012.05.021] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 05/17/2012] [Accepted: 05/23/2012] [Indexed: 12/20/2022]
|