51
|
Birder LA, Wolf-Johnston A, Wein AJ, Cheng F, Grove-Sullivan M, Kanai AJ, Watson AM, Stoltz D, Watkins SC, Robertson AM, Newman D, Dmochowski RR, Jackson EK. Purine nucleoside phosphorylase inhibition ameliorates age-associated lower urinary tract dysfunctions. JCI Insight 2020; 5:140109. [PMID: 32910805 PMCID: PMC7605521 DOI: 10.1172/jci.insight.140109] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/04/2020] [Indexed: 12/29/2022] Open
Abstract
In the aging population, lower urinary tract (LUT) dysfunction is common and often leads to storage and voiding difficulties classified into overlapping symptom syndromes. Despite prevalence and consequences of these syndromes, LUT disorders continue to be undertreated simply because there are few therapeutic options. LUT function and structure were assessed in aged (>25 months) male and female Fischer 344 rats randomized to oral treatment with a purine nucleoside phosphorylase (PNPase inhibitor) 8-aminoguanine (8-AG) or vehicle for 6 weeks. The bladders of aged rats exhibited multiple abnormalities: tactile insensitivity, vascular remodeling, reduced collagen-fiber tortuosity, increased bladder stiffness, abnormal smooth muscle morphology, swelling of mitochondria, and increases in urodamaging purine metabolites. Treatment of aged rats with 8-AG restored all evaluated histological, ultrastructural, and physiological abnormalities toward that of a younger state. 8-AG is an effective treatment that ameliorates key age-related structural and physiologic bladder abnormalities. Because PNPase inhibition blocks metabolism of inosine to hypoxanthine and guanosine to guanine, likely uroprotective effects of 8-AG are mediated by increased bladder levels of uroprotective inosine and guanosine and reductions in urodamaging hypoxanthine and xanthine. These findings demonstrate that 8-AG has translational potential for treating age-associated LUT dysfunctions and resultant syndromes in humans.
Collapse
Affiliation(s)
- Lori A Birder
- Department of Medicine, Renal-Electrolyte Division, and.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Alan J Wein
- Division of Urology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fangzhou Cheng
- Department of Mechanical Engineering and Materials Science, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mara Grove-Sullivan
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anthony J Kanai
- Department of Medicine, Renal-Electrolyte Division, and.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Alan M Watson
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Donna Stoltz
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anne M Robertson
- Department of Mechanical Engineering and Materials Science, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Diane Newman
- Division of Urology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Roger R Dmochowski
- Department of Urology, Vanderbilt Medical Center, Nashville, Tennessee, USA
| | - Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
52
|
Nindawat S, Agrawal V. Arabian Primrose leaf extract mediated synthesis of silver nanoparticles: their industrial and biomedical applications. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:1259-1271. [PMID: 33016155 DOI: 10.1080/21691401.2020.1817056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The present study encompasses green synthesis of silver nanoparticles (AgNPs) using aqueous leaf extract of Arabian Primrose within 6 min of reaction at 60 °C, pH 7 and their characterisation using physico-chemical analytical techniques. UV-Visible spectroscopy elucidated the surface plasmon resonance around 420 nm. FESEM and TEM images revealed that AgNPs were spherical with average diameter 10-60 nm. XRD pattern confirmed their crystalline nature. The leaf extract rich in phenolics and flavonoids was subjected to GC-MS analysis that identified bioactive compounds helping in reduction and stabilisation of AgNPs. The synthesised AgNPs possessed high anti-oxidant potential against DPPH and H2O2 radicals. Incidentally, the AgNPs acted as excellent nanocatalyst towards borohydride reduction and degradation of structurally different organic dyes. The AgNPs also exhibited selective colorimetric sensing of hazardous mercuric, ferric ions and ammonia. Such AgNPs were cytotoxic against HeLa cells (IC50 7.18 µg/mL) and compatible towards normal L20B cells. These AgNPs showed effective anti-microbial activity against different human pathogens tested (bacterial and fungal). This is probably the first report of AgNPs synthesis using Arabian Primrose leaf extract showing strong anti-oxidant, catalytic, biosensing, anti-cancer and anti-microbial activities and find remarkable applications in medical, industrial and ecological sectors.
Collapse
Affiliation(s)
- Shruti Nindawat
- Medicinal Plant Biotechnology Lab, Department of Botany, University of Delhi, Delhi, India
| | - Veena Agrawal
- Medicinal Plant Biotechnology Lab, Department of Botany, University of Delhi, Delhi, India
| |
Collapse
|
53
|
Camargo A, Dalmagro AP, M. Rosa J, B. Zeni AL, P. Kaster M, Tasca CI, S. Rodrigues AL. Subthreshold doses of guanosine plus ketamine elicit antidepressant-like effect in a mouse model of depression induced by corticosterone: Role of GR/NF-κB/IDO-1 signaling. Neurochem Int 2020; 139:104797. [DOI: 10.1016/j.neuint.2020.104797] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/01/2020] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
|
54
|
Guanosine modulates SUMO2/3-ylation in neurons and astrocytes via adenosine receptors. Purinergic Signal 2020; 16:439-450. [PMID: 32892251 PMCID: PMC7524998 DOI: 10.1007/s11302-020-09723-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
SUMOylation is a post-translational modification (PTM) whereby members of the Small Ubiquitin-like MOdifier (SUMO) family of proteins are conjugated to lysine residues in target proteins. SUMOylation has been implicated in a wide range of physiological and pathological processes, and much attention has been given to its role in neurodegenerative conditions. Due to its reported role in neuroprotection, pharmacological modulation of SUMOylation represents an attractive potential therapeutic strategy in a number of different brain disorders. However, very few compounds that target the SUMOylation pathway have been identified. Guanosine is an endogenous nucleoside with important neuromodulatory and neuroprotective effects. Experimental evidence has shown that guanosine can modulate different intracellular pathways, including PTMs. In the present study we examined whether guanosine alters global protein SUMOylation. Primary cortical neurons and astrocytes were treated with guanosine at 1, 10, 100, 300, or 500 μM at four time points, 1, 6, 24, or 48 h. We show that guanosine increases global SUMO2/3-ylation in neurons and astrocytes at 1 h at concentrations above 10 μM. The molecular mechanisms involved in this effect were evaluated in neurons. The guanosine-induced increase in global SUMO2/3-ylation was still observed in the presence of dipyridamole, which prevents guanosine internalization, demonstrating an extracellular guanosine-induced effect. Furthermore, the A1 adenosine receptor antagonist DPCPX abolished the guanosine-induced increase in SUMO2/3-ylation. The A2A adenosine receptor antagonist ZM241385 increased SUMOylation per se, but did not alter guanosine-induced SUMOylation, suggesting that guanosine may modulate SUMO2/3-ylation through an A1-A2A receptor interaction. Taken together, this is the first report to show guanosine as a SUMO2/3-ylation enhancer in astrocytes and neurons.
Collapse
|
55
|
Thomaz DT, Andreguetti RR, Binder LB, Scheffer DDL, Corrêa AW, Silva FRMB, Tasca CI. Guanosine Neuroprotective Action in Hippocampal Slices Subjected to Oxygen and Glucose Deprivation Restores ATP Levels, Lactate Release and Glutamate Uptake Impairment: Involvement of Nitric Oxide. Neurochem Res 2020; 45:2217-2229. [PMID: 32666283 DOI: 10.1007/s11064-020-03083-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
Stroke is a major cause of disability and death worldwide. Oxygen and glucose deprivation (OGD) in brain tissue preparations can reproduce several pathological features induced by stroke providing a valuable ex vivo protocol for studying the mechanism of action of neuroprotective agents. Guanosine, an endogenous guanine nucleoside, promotes neuroprotection in vivo and in vitro models of neurotoxicity. We previously showed that guanosine protective effect was mimicked by inhibition of nitric oxide synthases (NOS) activity. This study was designed to investigate the involvement of nitric oxide (NO) in the mechanisms related to the protective role of guanosine in rat hippocampal slices subjected to OGD followed by reoxygenation (OGD/R). Guanosine (100 μM) and the pan-NOS inhibitor, L-NAME (1 mM) afforded protection to hippocampal slices subjected to OGD/R. The presence of NO donors, DETA-NO (800 μM) or SNP (5 μM) increased reactive species production, and abolished the protective effect of guanosine or L-NAME against OGD/R. Guanosine or L-NAME treatment prevented the impaired ATP production, lactate release, and glutamate uptake following OGD/R. The presence of a NO donor also abolished the beneficial effects of guanosine or L-NAME on bioenergetics and glutamate uptake. These results showed, for the first time, that guanosine may regulate cellular bioenergetics in hippocampal slices subjected to OGD/R injury by a mechanism that involves the modulation of NO levels.
Collapse
Affiliation(s)
- Daniel Tonial Thomaz
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianópolis, SC, 88040-900, Brazil.,Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Rafaela Rafognatto Andreguetti
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Luisa Bandeira Binder
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianópolis, SC, 88040-900, Brazil.,Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Débora da Luz Scheffer
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianópolis, SC, 88040-900, Brazil.,Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Alisson Willms Corrêa
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Fátima Regina Mena Barreto Silva
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianópolis, SC, 88040-900, Brazil.,Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Carla Inês Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianópolis, SC, 88040-900, Brazil. .,Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, 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
|
56
|
Piermartiri TCB, Dos Santos B, Barros-Aragão FGQ, Prediger RD, Tasca CI. Guanosine Promotes Proliferation in Neural Stem Cells from Hippocampus and Neurogenesis in Adult Mice. Mol Neurobiol 2020; 57:3814-3826. [PMID: 32592125 DOI: 10.1007/s12035-020-01977-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 06/08/2020] [Indexed: 01/26/2023]
Abstract
Neural stem cells can generate new neurons in the mouse adult brain in a complex multistep process called neurogenesis. Several factors regulate this process, including neurotransmitters, hormones, neurotrophic factors, pharmacological agents, and environmental factors. Purinergic signaling, mainly the adenosinergic system, takes part in neurogenesis, being involved in cell proliferation, migration, and differentiation. However, the role of the purine nucleoside guanosine in neurogenesis remains unclear. Here, we examined the effect of guanosine by using the neurosphere assay derived from neural stem cells of adult mice. We found that continuous treatment with guanosine increased the number of neurospheres, neural stem cell proliferation, and neuronal differentiation. The effect of guanosine to increase the number of neurospheres was reduced by removing adenosine from the culture medium. We next traced the neurogenic effect of guanosine in vivo. The intraperitoneal treatment of adult C57BL/6 mice with guanosine (8 mg/kg) for 26 days increased the number of dividing bromodeoxyuridine (BrdU)-positive cells and also increased neurogenesis, as identified by measuring doublecortin (DCX)-positive cells in the dentate gyrus (DG) of the hippocampus. Antidepressant-like behavior in adult mice accompanied the guanosine-induced neurogenesis in the DG. These results provide new evidence of a pro-neurogenic effect of guanosine on neural stem/progenitor cells, and it was associated in vivo with antidepressant-like effects.
Collapse
Affiliation(s)
- Tetsade C B Piermartiri
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, UFSC, Florianópolis, SC, Brazil
| | - Beatriz Dos Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.,Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, UFSC, Florianópolis, SC, Brazil
| | | | - Rui D Prediger
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, UFSC, Florianópolis, SC, Brazil.,Departamento de Farmacologia, Centro de Ciências Biológicas, UFSC, Florianópolis, SC, Brazil
| | - Carla Inês Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil. .,Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, UFSC, Florianópolis, SC, Brazil.
| |
Collapse
|
57
|
Camargo A, Dalmagro AP, Zeni ALB, Rodrigues ALS. Guanosine potentiates the antidepressant-like effect of subthreshold doses of ketamine: Possible role of pro-synaptogenic signaling pathway. J Affect Disord 2020; 271:100-108. [PMID: 32479304 DOI: 10.1016/j.jad.2020.03.186] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/22/2020] [Accepted: 03/28/2020] [Indexed: 12/11/2022]
Abstract
Background Augmentation therapies may be effective strategies to potentiate the ketamine's actions with lower potential for knock-on effects. Thus, this study investigated the ability of combined administration of guanosine plus ketamine to elicit an antidepressant-like effect associated with mTOR pathway modulation. The ability of this combined administration to exert an antidepressant-like effect in a model of depression was also evaluated. Methods Mice were administered with subthreshold doses of ketamine (0.1 mg/kg, i.p.) and guanosine (0.01 mg/kg, p.o.) and submitted to the tail suspension test, and immunoblotting analyses (p-mTOR, p-p70S6K, PSD-95, GluA1, and synapsin) in the hippocampus and prefrontal cortex. The antidepressant-like effect of ketamine plus guanosine in mice subjected to administration of corticosterone (20 mg/kg, p.o., 21 days) was also evaluated. Results Ketamine plus guanosine treatment elicited an antidepressant-like effect, which was associated with increased mTOR (Ser2448) and p70S6K (Thr389) phosphorylation in the hippocampus, but not in the prefrontal cortex. Furthermore, increased PSD-95 and GluA1 immunocontent were observed in the prefrontal cortex, but not in the hippocampus of ketamine plus guanosine-treated mice. Reinforcing the notion that guanosine may potentiate the ketamine's behavioral response, a single administration of subthreshold doses of ketamine plus guanosine counteracted the corticosterone-induced depressive-like behavior. Conclusions Our results indicate that guanosine potentiates the antidepressant-like effect of subthreshold doses of ketamine, an effect likely associated with the stimulation of synaptogenic pathway in the hippocampus and prefrontal cortex, although with a different profile. The augmentation effect of ketamine by guanosine could have therapeutic relevance for patients with treatment-resistant depression.
Collapse
Affiliation(s)
- Anderson Camargo
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Ana Paula Dalmagro
- Department of Natural Sciences, Center of Exact and Natural Sciences, Universidade Regional de Blumenau, Blumenau, CEP 89030-903, Santa Catarina, Brazil
| | - Ana Lúcia B Zeni
- Department of Natural Sciences, Center of Exact and Natural Sciences, Universidade Regional de Blumenau, Blumenau, CEP 89030-903, Santa Catarina, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil.
| |
Collapse
|
58
|
Guanosine, a guanine-based nucleoside relaxed isolated corpus cavernosum from mice through cGMP accumulation. Purinergic Signal 2020; 16:241-249. [PMID: 32458299 DOI: 10.1007/s11302-020-09702-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 05/01/2020] [Indexed: 10/24/2022] Open
Abstract
In corpus cavernosum (CC), guanosine triphosphate (GTP) is converted into cyclic guanosine monophosphate (cGMP) to induce erection. The action of cGMP is terminated by phosphodiesterases and efflux transporters, which pump cGMP out of the cell. The nucleotides, GTP, and cGMP were detected in the extracellular space, and their hydrolysis lead to the formation of intermediate products, among them guanosine. Therefore, our study aims to pharmacologically characterize the effect of guanosine in isolated CC from mice. The penis was isolated and functional and biochemical analyses were carried out. The guanine-based nucleotides GTP, guanosine diphosphate, guanosine monophosphate, and cGMP relaxed mice corpus cavernosum, but the relaxation (90.7 ± 12.5%) induced by guanosine (0.000001-1 mM) was greater than that of the nucleotides (~ 45%, P < 0.05). Guanosine-induced relaxation was not altered in the presence of adenosine type 2A and 2B receptor antagonists. No augment was observed in the intracellular levels of cyclic adenosine monophosphate in tissues stimulated with guanosine. Inhibitors of nitric oxide synthase (L-NAME, 100 μM) and soluble guanylate cyclase (ODQ, 10 μM) produced a significant reduction in guanosine-induced relaxation in all concentrations studied, while in the presence of tadalafil (300 nM), a significant increase was observed. Pre-incubation of guanosine (100 μM) produced a 6.6-leftward shift in tadalafil-induced relaxation. The intracellular levels of cGMP were greater when CC was stimulated with guanosine. Inhibitors of ecto-nucleotidases and xanthine oxidase did not interfere in the response induced by guanosine. In conclusion, our study shows that guanosine relaxes mice CC and opens the possibility to test its role in models of erectile dysfunction.
Collapse
|
59
|
Almatarneh MH, Omeir RA, AL Demour S, Elayan IA, Islam S, Poirier RA. Hydrolytic deamination mechanisms of guanosine monophosphate: A computational study. COMPUT THEOR CHEM 2020. [DOI: 10.1016/j.comptc.2020.112732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
60
|
Mancinelli R, Fanò-Illic G, Pietrangelo T, Fulle S. Guanosine-Based Nucleotides, the Sons of a Lesser God in the Purinergic Signal Scenario of Excitable Tissues. Int J Mol Sci 2020; 21:ijms21051591. [PMID: 32111063 PMCID: PMC7084674 DOI: 10.3390/ijms21051591] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/28/2022] Open
Abstract
Purines are nitrogen compounds consisting mainly of a nitrogen base of adenine (ABP) or guanine (GBP) and their derivatives: nucleosides (nitrogen bases plus ribose) and nucleotides (nitrogen bases plus ribose and phosphate). These compounds are very common in nature, especially in a phosphorylated form. There is increasing evidence that purines are involved in the development of different organs such as the heart, skeletal muscle and brain. When brain development is complete, some purinergic mechanisms may be silenced, but may be reactivated in the adult brain/muscle, suggesting a role for purines in regeneration and self-repair. Thus, it is possible that guanosine-5′-triphosphate (GTP) also acts as regulator during the adult phase. However, regarding GBP, no specific receptor has been cloned for GTP or its metabolites, although specific binding sites with distinct GTP affinity characteristics have been found in both muscle and neural cell lines. Finally, even if the cross regulation mechanisms between the two different purines (ABP and GBP) are still largely unknown, it is now possible to hypothesize the existence of specific signal paths for guanosine-based nucleotides that are capable of modulating the intensity and duration of the intracellular signal, particularly in excitable tissues such as brain and muscle.
Collapse
Affiliation(s)
- Rosa Mancinelli
- Department of Neuroscience Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (R.M.); (T.P.)
- Interuniversity Institute of Miology (IIM), 66100 Chieti, Italy;
| | - Giorgio Fanò-Illic
- Interuniversity Institute of Miology (IIM), 66100 Chieti, Italy;
- Libera Università di Alcatraz, Santa Cristina di Gubbio, 06024 Gubbio, Italy
| | - Tiziana Pietrangelo
- Department of Neuroscience Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (R.M.); (T.P.)
- Interuniversity Institute of Miology (IIM), 66100 Chieti, Italy;
| | - Stefania Fulle
- Department of Neuroscience Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (R.M.); (T.P.)
- Interuniversity Institute of Miology (IIM), 66100 Chieti, Italy;
- Correspondence:
| |
Collapse
|
61
|
Kuś PM. Honey as Source of Nitrogen Compounds: Aromatic Amino Acids, Free Nucleosides and Their Derivatives. Molecules 2020; 25:molecules25040847. [PMID: 32075114 PMCID: PMC7070497 DOI: 10.3390/molecules25040847] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
The content of selected major nitrogen compounds including nucleosides and their derivatives was evaluated in 75 samples of seven varieties of honey (heather, buckwheat, black locust, goldenrod, canola, fir, linden) by targeted ultra-high performance liquid chromatography-diode array detector - high-resolution quadrupole time-of-flight mass spectrometry (UHPLC-DAD-QqTOF-MS) and determined by UHPLC-DAD. The honey samples contained nucleosides, nucleobases and their derivatives (adenine: 8.9 to 18.4 mg/kg, xanthine: 1.2 to 3.3 mg/kg, uridine: 17.5 to 51.2 mg/kg, guanosine: 2.0 to 4.1 mg/kg; mean amounts), aromatic amino acids (tyrosine: 7.8 to 263.9 mg/kg, phenylalanine: 9.5 to 64.1 mg/kg; mean amounts). The amounts of compounds significantly differed between some honey types. For example, canola honey contained a much lower amount of uridine (17.5 ± 3.9 mg/kg) than black locust where it was most abundant (51.2 ± 7.8 mg/kg). The presence of free nucleosides and nucleobases in different honey varieties is reported first time and supports previous findings on medicinal activities of honey reported in the literature as well as traditional therapy and may contribute for their explanation. This applies, e.g., to the topical application of honey in herpes infections, as well as its beneficial activity on cognitive functions as nootropic and neuroprotective, in neuralgia and is also important for the understanding of nutritional values of honey.
Collapse
Affiliation(s)
- Piotr M Kuś
- Department of Pharmacognosy and Herbal Medicines, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211a, 50-556 Wrocław, Poland
| |
Collapse
|
62
|
Zhang M, Jiang N, Chu Y, Postnikova O, Varghese R, Horvath A, Cheema AK, Golestaneh N. Dysregulated metabolic pathways in age-related macular degeneration. Sci Rep 2020; 10:2464. [PMID: 32051464 PMCID: PMC7016007 DOI: 10.1038/s41598-020-59244-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 01/27/2020] [Indexed: 01/03/2023] Open
Abstract
Age-related macular degeneration is a major cause of vision impairment in the Western world among people of 55 years and older. Recently we have shown that autophagy is dysfunctional in the retinal pigment epithelium (RPE) of the AMD donor eyes (AMD RPE). We also showed increased reactive oxygen (ROS) production, increased cytoplasmic glycogen accumulation, mitochondrial dysfunction and disintegration, and enlarged and annular LAMP-1-positive organelles in AMD RPE. However, the underlying mechanisms inducing these abnormalities remain to be elucidated. Here, by performing a comprehensive study, we show increased PAPR2 expression, deceased NAD+, and SIRT1, increased PGC-1α acetylation (inactive form), lower AMPK activity, and overactive mTOR pathway in AMD RPE as compared to normal RPE. Metabolomics and lipidomics revealed dysregulated metabolites in AMD RPE as compared to normal RPE, including glycerophospholipid metabolism, involved in autophagy, lipid, and protein metabolisms, glutathione, guanosine, and L-glutamic acid, which are implicated in protection against oxidative stress and neurotoxicity, further supporting our observations. Our data show dysregulated metabolic pathways as important contributors to AMD pathophysiology, and facilitate the development of new treatment strategies for this debilitating disease of the visual system.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Nisi Jiang
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Yi Chu
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Olga Postnikova
- Laboratory of Retinal Cell & Molecular Biology (HNW28), NIH/NEI, Bethesda, MD, 20814, USA
| | - Rency Varghese
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Anelia Horvath
- Department of Pharmacology and Physiology, Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC, 20037, USA
| | - Amrita K Cheema
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20057, USA.,Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Nady Golestaneh
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20057, USA. .,Department of Neurology, Georgetown University Medical Center, Washington, DC, 20057, USA. .,Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20057, USA.
| |
Collapse
|
63
|
Bast-Habersbrunner A, Fromme T. Purine Nucleotides in the Regulation of Brown Adipose Tissue Activity. Front Endocrinol (Lausanne) 2020; 11:118. [PMID: 32210919 PMCID: PMC7076073 DOI: 10.3389/fendo.2020.00118] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/24/2020] [Indexed: 01/06/2023] Open
Abstract
Non-shivering thermogenesis in mammalian brown adipose tissue is a powerful mechanism to defend normothermia in cold climates. To minimize the loss of chemical energy, the central functional component, mitochondrial uncoupling protein 1, UCP1, must be tightly regulated. The canonical pathway of UCP1 activation includes lipolytic release of free fatty acids in response to an adrenergic signal. Activating fatty acids overcome constitutive inhibition of UCP1 by the di- and triphosphate forms of purine nucleotides, i.e., ATP, ADP, GTP, and GDP. Cellular concentrations of inhibitory, free nucleotides are subject to significant, adrenergically induced alterations. The regulatory components involved may constitute novel drug targets to manipulate brown fat thermogenesis and thereby organismic energy balance. We here review evidence for and against a dominant role of nucleotides in thermogenic control, describe conceptual routes to endogenously and pharmacologically alter free nucleotide pool size, speculate on a signaling role of degradation products released from active brown fat, and highlight gaps in our understanding of signaling and metabolic pathways involved.
Collapse
Affiliation(s)
- Andrea Bast-Habersbrunner
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich, Germany
| | - Tobias Fromme
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich, Germany
- *Correspondence: Tobias Fromme
| |
Collapse
|
64
|
Lanznaster D, Massari CM, Marková V, Šimková T, Duroux R, Jacobson KA, Fernández-Dueñas V, Tasca CI, Ciruela F. Adenosine A 1-A 2A Receptor-Receptor Interaction: Contribution to Guanosine-Mediated Effects. Cells 2019; 8:E1630. [PMID: 31847113 PMCID: PMC6953045 DOI: 10.3390/cells8121630] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/04/2019] [Accepted: 12/11/2019] [Indexed: 01/02/2023] Open
Abstract
Guanosine, a guanine-based purine nucleoside, has been described as a neuromodulator that exerts neuroprotective effects in animal and cellular ischemia models. However, guanosine's exact mechanism of action and molecular targets have not yet been identified. Here, we aimed to elucidate a role of adenosine receptors (ARs) in mediating guanosine effects. We investigated the neuroprotective effects of guanosine in hippocampal slices from A2AR-deficient mice (A2AR-/-) subjected to oxygen/glucose deprivation (OGD). Next, we assessed guanosine binding at ARs taking advantage of a fluorescent-selective A2AR antagonist (MRS7396) which could engage in a bioluminescence resonance energy transfer (BRET) process with NanoLuc-tagged A2AR. Next, we evaluated functional AR activation by determining cAMP and calcium accumulation. Finally, we assessed the impact of A1R and A2AR co-expression in guanosine-mediated impedance responses in living cells. Guanosine prevented the reduction of cellular viability and increased reactive oxygen species generation induced by OGD in hippocampal slices from wild-type, but not from A2AR-/- mice. Notably, while guanosine was not able to modify MRS7396 binding to A2AR-expressing cells, a partial blockade was observed in cells co-expressing A1R and A2AR. The relevance of the A1R and A2AR interaction in guanosine effects was further substantiated by means of functional assays (i.e., cAMP and calcium determinations), since guanosine only blocked A2AR agonist-mediated effects in doubly expressing A1R and A2AR cells. Interestingly, while guanosine did not affect A1R/A2AR heteromer formation, it reduced A2AR agonist-mediated cell impedance responses. Our results indicate that guanosine-induced effects may require both A1R and A2AR co-expression, thus identifying a molecular substrate that may allow fine tuning of guanosine-mediated responses.
Collapse
Affiliation(s)
- Débora Lanznaster
- Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, Brazil;
| | - Caio M. Massari
- Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, Brazil;
| | - Vendula Marková
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, 08907 L’Hospitalet de Llobregat, Spain; (V.M.); (T.Š.)
- Institut de Neurociències, Universitat de Barcelona, 08035 Barcelona, Spain
| | - Tereza Šimková
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, 08907 L’Hospitalet de Llobregat, Spain; (V.M.); (T.Š.)
- Institut de Neurociències, Universitat de Barcelona, 08035 Barcelona, Spain
| | - Romain Duroux
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (R.D.); (K.A.J.)
| | - Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (R.D.); (K.A.J.)
| | - Víctor Fernández-Dueñas
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, 08907 L’Hospitalet de Llobregat, Spain; (V.M.); (T.Š.)
- Institut de Neurociències, Universitat de Barcelona, 08035 Barcelona, Spain
| | - Carla I. Tasca
- Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, Brazil;
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, Brazil
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, 08907 L’Hospitalet de Llobregat, Spain; (V.M.); (T.Š.)
- Institut de Neurociències, Universitat de Barcelona, 08035 Barcelona, Spain
| |
Collapse
|
65
|
Dal-Cim T, Poluceno GG, Lanznaster D, de Oliveira KA, Nedel CB, Tasca CI. Guanosine prevents oxidative damage and glutamate uptake impairment induced by oxygen/glucose deprivation in cortical astrocyte cultures: involvement of A 1 and A 2A adenosine receptors and PI3K, MEK, and PKC pathways. Purinergic Signal 2019; 15:465-476. [PMID: 31520282 DOI: 10.1007/s11302-019-09679-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/22/2019] [Indexed: 12/31/2022] Open
Abstract
Glial cells are involved in multiple cerebral functions that profoundly influence brain tissue viability during ischemia, and astrocytes are the main source of extracellular purines as adenosine and guanosine. The endogenous guanine-based nucleoside guanosine is a neuromodulator implicated in important processes in the brain, such as modulation of glutamatergic transmission and protection against oxidative and inflammatory damage. We evaluated if the neuroprotective effect of guanosine is also observed in cultured cortical astrocytes subjected to oxygen/glucose deprivation (OGD) and reoxygenation. We also assessed the involvement of A1 and A2A adenosine receptors and phosphatidylinositol-3 kinase (PI3K), MAPK, and protein kinase C (PKC) signaling pathways on the guanosine effects. OGD/reoxygenation decreased cell viability and glutamate uptake and increased reactive oxygen species (ROS) production in cultured astrocytes. Guanosine treatment prevented these OGD-induced damaging effects. Dipropyl-cyclopentyl-xanthine (an adenosine A1 receptor antagonist) and 4-[2-[[6-amino-9-(N-ethyl-β-D-ribofuranuronamidosyl)-9H-purin-2-yl]amino]ethyl] benzenepropanoic acid hydrochloride (an adenosine A2A receptor agonist) abolished guanosine-induced protective effects on ROS production, glutamate uptake, and cell viability. The PI3K pathway inhibitor 2-morpholin-4-yl-8-phenylchromen-4-one, the extracellular-signal regulated kinase kinase (MEK) inhibitor 2'-amino-3'-methoxyflavone, or the PKC inhibitor chelerythrine abolished the guanosine effect of preventing OGD-induced cells viability reduction. PI3K inhibition partially prevented the guanosine effect of reducing ROS production, whereas MEK and PKC inhibitions prevented the guanosine effect of restoring glutamate uptake. The total immunocontent of the main astrocytic glutamate transporter glutamate transporter-1 (GLT-1) was not altered by OGD and guanosine. However, MEK and PKC inhibitions also abolished the guanosine effect of increasing cell-surface expression of GLT-1 in astrocytes subjected to OGD. Then, guanosine prevents oxidative damage and stimulates astrocytic glutamate uptake during ischemic events via adenosine A1 and A2A receptors and modulation of survival signaling pathways, contributing to microenvironment homeostasis that culminates in neuroprotection.
Collapse
Affiliation(s)
- Tharine Dal-Cim
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianopolis, SC, 88040-900, Brazil
- Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| | - Gabriela G Poluceno
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianopolis, SC, 88040-900, Brazil
- Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| | - Débora Lanznaster
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianopolis, SC, 88040-900, Brazil
- Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| | - Karen A de Oliveira
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianopolis, SC, 88040-900, Brazil
- Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| | - Claudia B Nedel
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| | - Carla I Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, Florianopolis, SC, 88040-900, Brazil.
- Programa de Pós-graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil.
- Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil.
| |
Collapse
|
66
|
Decker H, Piermartiri TCB, Nedel CB, Romão LF, Francisco SS, Dal-Cim T, Boeck CR, Moura-Neto V, Tasca CI. Guanosine and GMP increase the number of granular cerebellar neurons in culture: dependence on adenosine A 2A and ionotropic glutamate receptors. Purinergic Signal 2019; 15:439-450. [PMID: 31478180 DOI: 10.1007/s11302-019-09677-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
The guanine-based purines (GBPs) have essential extracellular functions such as modulation of glutamatergic transmission and trophic effects on neurons and astrocytes. We previously showed that GBPs, such as guanosine-5'-monophosphate (GMP) or guanosine (GUO), promote the reorganization of extracellular matrix proteins in astrocytes, and increase the number of neurons in a neuron-astrocyte co-culture protocol. To delineate the molecular basis underlying these effects, we isolated cerebellar neurons in culture and treated them with a conditioned medium derived from astrocytes previously exposed to GUO or GMP (GBPs-ACM) or, directly, with GUO or GMP. Agreeing with the previous studies, there was an increase in the number of β-tubulin III-positive neurons in both conditions, compared with controls. Interestingly, the increase in the number of neurons in the neuronal cultures treated directly with GUO or GMP was more prominent, suggesting a direct interaction of GBPs on cerebellar neurons. To investigate this issue, we assessed the role of adenosine and glutamate receptors and related intracellular signaling pathways after GUO or GMP treatment. We found an involvement of A2A adenosine receptors, ionotropic glutamate N-methyl-D-aspartate (NMDA), and non-NMDA receptors in the increased number of cerebellar neurons. The signaling pathways extracellular-regulated kinase (ERK), calcium-calmodulin-dependent kinase-II (CaMKII), protein kinase C (PKC), phosphatidilinositol-3'-kinase (PI3-K), and protein kinase A (PKA) are also potentially involved with GMP and GUO effect. Such results suggest that GMP and GUO, and molecules released in GBPs-ACM promote the survival or maturation of primary cerebellar neurons or both via interaction with adenosine and glutamate receptors.
Collapse
Affiliation(s)
- Helena Decker
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Tetsade C B Piermartiri
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
| | - Cláudia B Nedel
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
| | - Luciana F Romão
- Departamento de Anatomia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brasil
| | - Sheila S Francisco
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
| | - Tharine Dal-Cim
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil
| | - Carina R Boeck
- Programa de Pós-graduação em Nanociências, Universidade Franciscana, Santa Maria, RS, Brasil
| | - Vivaldo Moura-Neto
- Departamento de Anatomia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brasil
- Instituto Estadual do Cérebro Paulo Niemeyer da Secretaria de Estado de Saúde do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Carla I Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900, Florianópolis, SC, Brasil.
| |
Collapse
|
67
|
Liska A, Triskova I, Ludvik J, Trnkova L. Oxidation potentials of guanine, guanosine and guanosine-5′-monophosphate: Theory and experiment. Electrochim Acta 2019. [DOI: 10.1016/j.electacta.2019.06.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
68
|
Zhu H, Chandra A, Geng X, Cheng Z, Tong Y, Du H, Ding Y. Low dose concomitant treatment with chlorpromazine and promethazine is safe in acute ischemic stroke. J Neurosurg Sci 2019; 63:265-269. [DOI: 10.23736/s0390-5616.19.04665-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
69
|
Zhu H, Geng X, Rajah GB, Fu P, Du H. Brain ultrasound for diagnosis and prognosis in the neurological intensive care unit: a mini review for current development. Neurol Res 2019; 41:691-696. [PMID: 31030634 DOI: 10.1080/01616412.2019.1609158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Haomeng Zhu
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University at the Detroit Medical Center, Detroit MI, USA
| | - Gary B. Rajah
- Department of Neurosurgery, Wayne State University at the Detroit Medical Center, Detroit MI, USA
| | - Paul Fu
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Huishan Du
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
70
|
A 1 rather than A 2A adenosine receptor as a possible target of Guanosine effects on mitochondrial dysfunction following Traumatic Brain Injury in rats. Neurosci Lett 2019; 704:141-144. [PMID: 30974229 DOI: 10.1016/j.neulet.2019.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 03/27/2019] [Accepted: 04/06/2019] [Indexed: 11/21/2022]
Abstract
Traumatic brain injury (TBI) represents one of the leading causes of death worldwide. Its pathophysiology involves several neurochemical events including mitochondrial dysfunction. Since mitochondrial respiration plays a key role in cell survival, pharmacological interventions targeting mitochondrial function have been highlighted as a powerful tool against the neurodegenerative process triggered by TBI. Guanosine (GUO), a neuroprotective molecule in different neurological disorders involving neurotoxicity, has shown protective properties after TBI, however its mechanism of action is not well understood in the central nervous system (CNS). Therefore, the aim of this study is to evaluate the possible target receptor involved in the protective GUO effects on TBI-induced mitochondrial dysfunction in the cerebral cortex of rats. Results show that a single dose of GUO (7.5 mg/kg) injected 40 min after a fluid percussion injury (FPI) protects against loss of mitochondrial membrane potential and increase of reactive oxygen species 8 h post-TBI. These effects were specifically blocked by a pretreatment (10 min after TBI) with an A1 adenosine receptor antagonist (DPCPX 1 mg/kg). In contrast, pretreatment with an A2A adenosine receptor antagonist (SCH 58261 0.05 mg/kg) did not alter GUO effects. These findings suggest that acute GUO neuroprotection following TBI involves the modulation of the adenosinergic system, especially A1 adenosine receptor.
Collapse
|
71
|
Cryan MT, Ross AE. Scalene Waveform for Codetection of Guanosine and Adenosine Using Fast-Scan Cyclic Voltammetry. Anal Chem 2019; 91:5987-5993. [DOI: 10.1021/acs.analchem.9b00450] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Michael T. Cryan
- Department of Chemistry, University of Cincinnati, 312 College Drive, 404 Crosley Tower, Cincinnati, Ohio 45221-0172, United States
| | - Ashley E. Ross
- Department of Chemistry, University of Cincinnati, 312 College Drive, 404 Crosley Tower, Cincinnati, Ohio 45221-0172, United States
| |
Collapse
|
72
|
Kallassy H, Fayyad-Kazan H, Makki R, Kaeen M, Sakr A, Alyamani O, El Dirani R, Hamade E, Fayyad-Kazan M, Badran B. Chemical Composition and Biological Activities of Lebanese Pentapera Plant. Med Sci Monit Basic Res 2019; 25:88-99. [PMID: 30867404 PMCID: PMC6431113 DOI: 10.12659/msmbr.914741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Due to their chemical constituents and biological properties, plants have long been used to control life-threatening diseases. The flora of Lebanon includes many plants that have already been demonstrated to have medicinal value, and other species, such as Pentapera sicula libanotica, that are yet to be characterized. The present study characterized the chemical composition, anti-oxidant, anti-inflammatory, and anti-proliferative potential of aqueous, ethanol, and methanol extracts derived from the leaves of the Lebanese Pentapera plant. Material/Methods High-performance liquid chromatography (HPLC) was used to determine the chemical composition. Gas chromatography (GC) coupled with mass spectrometry (MS) was applied to determine the content of essential oil. DPPH radical scavenging assay was performed to evaluate the anti-oxidant potential. The anti-inflammatory potential was assessed using quantitative real-time PCR (qRT-PCR) by measuring TNF-α, IL-6, and CCL4 mRNA levels, and we assessed Cox-2 and iNOS proteins levels using Western blot (WB) analysis. MTT assay was carried out to determine the anti-proliferative potential. Results We identified, mainly in the alcoholic (methanol and ethanol) extracts, distinct bioactive compounds with pharmacological relevance. In parallel, with their phytochemical content, these 2 extracts showed significant anti-oxidant, anti-inflammatory and anti-proliferative capacities. Conclusion Pentapera sicula libanotica appears to be a promising pharmacological tool.
Collapse
Affiliation(s)
- Hany Kallassy
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Rawan Makki
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Monya Kaeen
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Amer Sakr
- Toxicology Laboratory, Faculty of Health Sciences, American University of Science and Technology, Beirut, Lebanon
| | - Osama Alyamani
- Toxicology Laboratory, Faculty of Health Sciences, American University of Science and Technology, Beirut, Lebanon
| | - Rim El Dirani
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Eva Hamade
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Mohammad Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| |
Collapse
|
73
|
Xu J, Patassini S, Rustogi N, Riba-Garcia I, Hale BD, Phillips AM, Waldvogel H, Haines R, Bradbury P, Stevens A, Faull RLM, Dowsey AW, Cooper GJS, Unwin RD. Regional protein expression in human Alzheimer's brain correlates with disease severity. Commun Biol 2019; 2:43. [PMID: 30729181 PMCID: PMC6361956 DOI: 10.1038/s42003-018-0254-9] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 12/03/2018] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that currently affects 36 million people worldwide with no effective treatment available. Development of AD follows a distinctive pattern in the brain and is poorly modelled in animals. Therefore, it is vital to widen the spatial scope of the study of AD and prioritise the study of human brains. Here we show that functionally distinct human brain regions display varying and region-specific changes in protein expression. These changes provide insights into the progression of disease, novel AD-related pathways, the presence of a gradient of protein expression change from less to more affected regions and a possibly protective protein expression profile in the cerebellum. This spatial proteomics analysis provides a framework which can underpin current research and open new avenues to enhance molecular understanding of AD pathophysiology, provide new targets for intervention and broaden the conceptual frameworks for future AD research.
Collapse
Affiliation(s)
- Jingshu Xu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, Private Bag 92019, Auckland, 1142 New Zealand
| | - Stefano Patassini
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, Private Bag 92019, Auckland, 1142 New Zealand
| | - Nitin Rustogi
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
| | - Isabel Riba-Garcia
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
| | - Benjamin D. Hale
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
| | - Alexander M Phillips
- Department of Electrical Engineering and Electronics, University of Liverpool, Liverpool, L69 3GJ UK
| | - Henry Waldvogel
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142 New Zealand
| | - Robert Haines
- Research IT, The University of Manchester, Manchester, M13 9PL UK
| | - Phil Bradbury
- Research IT, The University of Manchester, Manchester, M13 9PL UK
| | - Adam Stevens
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PL UK
| | - Richard L. M. Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142 New Zealand
| | - Andrew W. Dowsey
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
- Department of Population Health Sciences and Bristol Veterinary School, Faculty of Health Sciences, University of Bristol, Bristol, BS8 2BN UK
| | - Garth J. S. Cooper
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, Private Bag 92019, Auckland, 1142 New Zealand
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142 New Zealand
| | - Richard D. Unwin
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
| |
Collapse
|
74
|
Guanosine protects against Ca 2+-induced mitochondrial dysfunction in rats. Biomed Pharmacother 2019; 111:1438-1446. [PMID: 30841459 DOI: 10.1016/j.biopha.2019.01.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 01/11/2023] Open
Abstract
Mitochondria play an important role in cell life and in the regulation of cell death. In addition, mitochondrial dysfunction contributes to a wide range of neuropathologies. The nucleoside Guanosine (GUO) is an endogenous molecule, presenting antioxidant properties, possibly due to its direct scavenging ability and/or from its capacity to activate the antioxidant defense system. GUO demonstrate a neuroprotective effect due to the modulation of the glutamatergic system and maintenance of the redox system. Thus, considering the few studies focused on the direct effects of GUO on mitochondrial bioenergetics, we designed a study to evaluate the in vitro effects of GUO on rat mitochondrial function, as well as against Ca2+-induced impairment. Our results indicate that GUO prevented mitochondrial dysfunction induced by Ca2+ misbalance, once GUO was able to reduce mitochondrial swelling in the presence of Ca2+, as well as ROS production and hydrogen peroxide levels, and to increase manganese superoxide dismutase activity, oxidative phosphorylation and tricarboxylic acid cycle activities. Our study indicates for the first time that GUO could direct prevent the mitochondrial damage induced by Ca2+ and that these effects were not related to its scavenging properties. Our data indicates that GUO could be included as a new pharmacological strategy for diseases linked to mitochondrial dysfunction.
Collapse
|
75
|
Cryan MT, Ross AE. Subsecond detection of guanosine using fast-scan cyclic voltammetry. Analyst 2019; 144:249-257. [PMID: 30484441 DOI: 10.1039/c8an01547c] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Guanosine is an important neuromodulator and neuroprotector in the brain and is involved in many pathological conditions, including ischemia and neuroinflammation. Traditional methods to detect guanosine in the brain, like HPLC, offer low limits of detection and are robust; however, subsecond detection is not possible. Here, we present a method for detecting rapid fluctuations of guanosine concentration in real-time using fast-scan cyclic voltammetry (FSCV) at carbon-fiber microelectrodes. The optimized waveform scanned from -0.4 V to 1.3 V and back at a rate of 400 V s-1 and application frequency of 10 Hz. Potential limits were chosen to increase selectivity of guanosine over the structurally similar interferent adenosine. Two oxidation peaks were detected with the optimized waveform: the primary oxidation reaction occurred at 1.3 V and the secondary oxidation at 0.8 V. Guanosine detection was stable over time with a limit of detection of 30 ± 10 nM, which permits its use to monitor low nanomolar fluctuations in the brain. To demonstrate the feasibility of the method for in-tissue detection, guanosine was exogenously applied and detected within live rat brain slices. This paper demonstrates the first characterization of guanosine using FSCV, and will be a valuable method for measuring signaling dynamics during guanosine neuromodulation and protection.
Collapse
Affiliation(s)
- Michael T Cryan
- University of Cincinnati, Department of Chemistry, 312 College Dr., 404 Crosley Tower, Cincinnati, OH 45221-0172, USA.
| | - Ashley E Ross
- University of Cincinnati, Department of Chemistry, 312 College Dr., 404 Crosley Tower, Cincinnati, OH 45221-0172, USA.
| |
Collapse
|
76
|
Camargo A, Rodrigues ALS. Novel Targets for Fast Antidepressant Responses: Possible Role of Endogenous Neuromodulators. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2019; 3:2470547019858083. [PMID: 32440595 PMCID: PMC7219953 DOI: 10.1177/2470547019858083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022]
Abstract
The available medications for the treatment of major depressive disorder have limitations, particularly their limited efficacy, delayed therapeutic effects, and the side effects associated with treatment. These issues highlight the need for better therapeutic agents that provide more efficacious and faster effects for the management of this disorder. Ketamine, an N-methyl-D-aspartate receptor antagonist, is the prototype for novel glutamate-based antidepressants that has been shown to cause a rapid and sustained antidepressant effect even in severe refractory depressive patients. Considering the importance of these findings, several studies have been conducted to elucidate the molecular targets for ketamine's effect. In addition, efforts are under way to characterize ketamine-like drugs. This review focuses particularly on evidence that endogenous glutamatergic neuromodulators may be able to modulate mood and to elicit fast antidepressant responses. Among these molecules, agmatine and creatine stand out as those with more published evidence of similarities with ketamine, but guanosine and ascorbic acid have also provided promising results. The possibility that these neuromodulators and ketamine have common neurobiological mechanisms, mainly the ability to activate mechanistic target of rapamycin and brain-derived neurotrophic factor signaling, and synthesis of synaptic proteins in the prefrontal cortex and/or hippocampus is presented and discussed.
Collapse
Affiliation(s)
- Anderson Camargo
- Neuroscience Postgraduate Program,
Center of Biological Sciences, Universidade Federal de Santa Catarina,
Florianópolis, Brazil
| | - Ana Lúcia S. Rodrigues
- Department of Biochemistry, Center of
Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis,
Brazil
| |
Collapse
|
77
|
Marques NF, Massari CM, Tasca CI. Guanosine Protects Striatal Slices Against 6-OHDA-Induced Oxidative Damage, Mitochondrial Dysfunction, and ATP Depletion. Neurotox Res 2018; 35:475-483. [PMID: 30417317 DOI: 10.1007/s12640-018-9976-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 01/18/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by loss of dopaminergic neurons in substantia nigra pars compacta which induces severe motor symptoms. 6-OHDA is a neurotoxin widely used in PD animal models due to its high affinity by dopamine transporter, its rapid non-enzymatic auto-oxidation which generates reactive oxygen species (ROS), oxidative stress, and for induced mitochondrial dysfunction. We previously reported an in vitro protocol of 6-OHDA-induced toxicity in brain regions slices, as a simple and sensitive assay to screen for protective compounds related to PD. Guanosine (GUO), a guanine-based purine nucleoside, is a neuroprotective molecule that is showing promising effects as an antiparkinsonian agent. To investigate the mechanisms involved on GUO-induced neuroprotection, slices of cortex, striatum, and hippocampus were incubated with GUO in the presence of 6-OHDA (100 μM). 6-OHDA promoted a decrease in cellular viability and increased ROS generation in all brain regions. Disruption of mitochondrial potential, depletion in intracellular ATP levels, and increase in cell membrane permeabilization were evidenced in striatal slices. GUO prevented the increase in ROS generation, disruption in mitochondrial potential, and depletion of intracellular ATP induced by 6-OHDA in striatal slices. In conclusion, GUO was effective to prevent oxidative events before cell damage, such as mitochondrial disruption, intracellular ATP levels depletion, and ROS generation in striatal slices subjected to in vitro 6-OHDA-induced toxicity.
Collapse
Affiliation(s)
- Naiani Ferreira Marques
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Caio Marcos Massari
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Carla Inês Tasca
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil. .,Departamento de Bioquímica, CCB, UFSC, Trindade, Florianópolis, Santa Catarina, 88040-900, Brazil.
| |
Collapse
|
78
|
Tasca CI, Lanznaster D, Oliveira KA, Fernández-Dueñas V, Ciruela F. Neuromodulatory Effects of Guanine-Based Purines in Health and Disease. Front Cell Neurosci 2018; 12:376. [PMID: 30459558 PMCID: PMC6232889 DOI: 10.3389/fncel.2018.00376] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 10/02/2018] [Indexed: 12/18/2022] Open
Abstract
The function of guanine-based purines (GBPs) is mostly attributed to the intracellular modulation of heteromeric and monomeric G proteins. However, extracellular effects of guanine derivatives have also been recognized. Thus, in the central nervous system (CNS), a guanine-based purinergic system that exerts neuromodulator effects, has been postulated. The thesis that GBPs are neuromodulators emerged from in vivo and in vitro studies, in which neurotrophic and neuroprotective effects of these kinds of molecules (i.e., guanosine) were demonstrated. GBPs induce several important biological effects in rodent models and have been shown to reduce seizures and pain, stabilize mood disorder behavior and protect against gliomas and diseases related with aging, such as ischemia or Parkinson and Alzheimer diseases. In vitro studies to evaluate the protective and trophic effects of guanosine, and of the nitrogenous base guanine, have been fundamental for understanding the mechanisms of action of GBPs, as well as the signaling pathways involved in their biological roles. Conversely, although selective binding sites for guanosine have been identified in the rat brain, GBP receptors have not been still described. In addition, GBP neuromodulation may depend on the capacity of GBPs to interact with well-known membrane proteins in glutamatergic and adenosinergic systems. Overall, in this review article, we present up-to-date GBP biology, focusing mainly on the mechanisms of action that may lead to the neuromodulator role of GBPs observed in neurological disorders.
Collapse
Affiliation(s)
- Carla I Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Débora Lanznaster
- Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,UMR 1253, Team 2, INSERM/University of Tours, Tours, France
| | - Karen A Oliveira
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Victor Fernández-Dueñas
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
79
|
Preventive effects of guanosine on intestinal inflammation in 2, 4-dinitrobenzene sulfonic acid (DNBS)-induced colitis in rats. Inflammopharmacology 2018; 27:349-359. [PMID: 29907915 DOI: 10.1007/s10787-018-0506-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/06/2018] [Indexed: 02/08/2023]
|
80
|
Teixeira LV, Almeida RF, Rohden F, Martins LAM, Spritzer PM, de Souza DOG. Neuroprotective Effects of Guanosine Administration on In Vivo Cortical Focal Ischemia in Female and Male Wistar Rats. Neurochem Res 2018; 43:1476-1489. [PMID: 29855847 DOI: 10.1007/s11064-018-2562-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/23/2018] [Accepted: 05/26/2018] [Indexed: 02/07/2023]
Abstract
Guanosine (GUO) has neuroprotective effects in experimental models of brain diseases involving glutamatergic excitotoxicity in male animals; however, its effects in female animals are poorly understood. Thus, we investigated the influence of gender and GUO treatment in adult male and female Wistar rats submitted to focal permanent cerebral ischemia in the motor cortex brain. Female rats were subdivided into non-estrogenic and estrogenic phase groups by estrous cycle verification. Immediately after surgeries, the ischemic animals were treated with GUO or a saline solution. Open field and elevated plus maze tasks were conducted with ischemic and naïve animals. Cylinder task, immunohistochemistry and infarct volume analyses were conducted only with ischemic animals. Female GUO groups achieved a full recovery of the forelimb symmetry at 28-35 days after the insult, while male GUO groups only partially recovered at 42 days, in the final evaluation. The ischemic insult affected long-term memory habituation to novelty only in female groups. Anxiety-like behavior, astrocyte morphology and infarct volume were not affected. Regardless the estrous cycle, the ischemic injury affected differently female and male animals. Thus, this study points that GUO is a potential neuroprotective compound in experimental stroke and that more studies, considering the estrous cycle, with both genders are recommended in future investigation concerning brain diseases.
Collapse
Affiliation(s)
- Luciele Varaschini Teixeira
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Roberto Farina Almeida
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Francieli Rohden
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Leo Anderson Meira Martins
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Poli Mara Spritzer
- Department of Physiology, Laboratory of Molecular Endocrinology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Diogo Onofre Gomes de Souza
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
81
|
Shinohara Y, Tsukimoto M. Guanine and inosine nucleotides/nucleosides suppress murine T cell activation. Biochem Biophys Res Commun 2018. [PMID: 29524424 DOI: 10.1016/j.bbrc.2018.03.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Damaged tissues and cells release intracellular purine nucleotides, which serve as intercellular signaling factors. We previously showed that exogenously added adenine nucleotide (250 μM ATP) suppressed the activation of murine splenic T lymphocytes. Here, we examined the effects of other purine nucleotides/nucleosides on mouse T cell activation. First, we found that pretreatment of mouse spleen T cells with 250 μM GTP, GDP, GMP, guanosine, ITP, IDP, IMP or inosine significantly reduced the release of stimulus-inducible cytokine IL-2. This suppression of IL-2 release was not caused by induction of cell death. Further studies with GTP, ITP, guanosine and inosine showed that pretreatment with these nucleotides/nucleosides also suppressed release of IL-6. However, these nucleotides/nucleosides did not suppress stimulus-induced phosphorylation of ERK1/2, suggesting that the suppression of the release of inflammatory cytokines does not involve inhibition of ERK1/2 signaling. In contrast to ATP pretreatment at the same concentration, guanine or inosine nucleotides/nucleosides did not attenuate the expression of CD25. Our findings indicate that exogenous guanine or inosine nucleotides/nucleosides can suppress inflammatory cytokine release from T cells, and may be promising candidates for use as supplementary agents in the treatment of T cell-mediated immune diseases.
Collapse
Affiliation(s)
- Yuria Shinohara
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda-shi, Chiba, Japan
| | - Mitsutoshi Tsukimoto
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda-shi, Chiba, Japan.
| |
Collapse
|
82
|
Zuccarini M, Giuliani P, Frinchi M, Mudò G, Serio RM, Belluardo N, Buccella S, Carluccio M, Condorelli DF, Caciagli F, Ciccarelli R, Di Iorio P. Uncovering the Signaling Pathway behind Extracellular Guanine-Induced Activation of NO System: New Perspectives in Memory-Related Disorders. Front Pharmacol 2018; 9:110. [PMID: 29515443 PMCID: PMC5826394 DOI: 10.3389/fphar.2018.00110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/31/2018] [Indexed: 12/31/2022] Open
Abstract
Mounting evidence suggests that the guanine-based purines stand out as key player in cell metabolism and in several models of neurodegenerative disorders, such as Parkinson's and Alzheimer's diseases. Guanosine (GUO) and guanine (GUA) are extracellular signaling molecules derived from the breakdown of the correspondent nucleotide, GTP, and their intracellular and extracellular levels are regulated by the fine-tuned activity of two major enzymes, purine nucleoside phosphorylase (PNP) and guanine deaminase (GDA). Noteworthy, GUO and GUA, seem to play opposite roles in the modulation of cognitive functions, such as learning and memory. Indeed GUO, despite exerting neuroprotective, anti-apoptotic and neurotrophic effects, causes a decay of cognitive activities, whereas GUA administration in rats results in working memory improvement (prevented by L-NAME pre-treatment). This study was designed to investigate, in a model of SH-SY5Y neuroblastoma cell line, the signal transduction pathway activated by extracellular GUA. Altogether, our results showed that: (i) in addition to an enhanced phosphorylation of ASK1, p38 and JNK, likely linked to a non-massive and transient ROS production, the PKB/NO/sGC/cGMP/PKG/ERK cascade seems to be the main signaling pathway elicited by extracellular GUA; (ii) the activation of this pathway occurs in a pertussis-toxin sensitive manner, thus suggesting the involvement of a putative G protein coupled receptor; (iii) the GUA-induced NO production, strongly reduced by cell pre-treatment with L-NAME, is negatively modulated by the EPAC-cAMP-CaMKII pathway, which causes the over-expression of GDA that, in turn, reduces the levels of GUA. These molecular mechanisms activated by GUA may be useful to support our previous observation showing that GUA improves learning and memory functions through the stimulation of NO signaling pathway, and underscore the therapeutic potential of oral administration of guanine for treating memory-related disorders.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Monica Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Rosa Maria Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Silvana Buccella
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | | | - Francesco Caciagli
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| |
Collapse
|
83
|
Chen F, He Y, Wang P, Wei P, Feng H, Rao Y, Shi J, Tian J. Banxia Xiexin decoction ameliorated cognition via the regulation of insulin pathways and glucose transporters in the hippocampus of APPswe/PS1dE9 mice. Int J Immunopathol Pharmacol 2018; 32:2058738418780066. [PMID: 29873261 PMCID: PMC5992807 DOI: 10.1177/2058738418780066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 05/07/2018] [Indexed: 01/09/2023] Open
Abstract
Reduced glucose utilization and deficient energy metabolism that occur in the early stages of Alzheimer's disease correlate with impaired cognition, and this information is evidence that Alzheimer's disease is a metabolic disease that is associated with brain insulin/insulin-like growth factor resistance. This research aimed to investigate the effects of Banxia Xiexin decoction (BXD) on cognitive deficits in APPswe/PS1dE9 double transgenic mice and verify the hypothesis that BXD treatment improves cognitive function via improving insulin signalling, glucose metabolism and synaptic plasticity in the hippocampus of APPswe/PS1dE9 double transgenic mice. We used 3-month-old APPswe/PS1dE9 double transgenic mice as the case groups and wild-type littermates of the double transgenic mice from the same colony as the control group. Forty-five APPswe/PS1dE9 double transgenic mice were randomly divided into the model group, donepezil group and BXD group. The mice in the control and model groups were administered 0.5% carboxymethyl cellulose orally. The Morris water maze and step-down test were conducted to evaluate the cognitive performance of APPswe/PS1dE9 double transgenic mice after BXD treatment. Ultrastructure of synapses was observed in the hippocampal CA1 area. Proteins involved in insulin signalling pathways and glucose transports in the hippocampus were assessed through immunohistochemical staining and western blot. After 3 months intervention, we found that BXD treatment improved cognitive performance and the synaptic quantity and ultrastructure, restored insulin signalling and increased the expression of glucose transporter 1 (GLUT1) and GLUT3 levels. These findings suggest that the beneficial effect of BXD on cognition may be due to the improvement of insulin signalling, glucose metabolism and synaptic plasticity.
Collapse
Affiliation(s)
- Fang Chen
- Key Laboratory of Chinese Internal
Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
- Key Laboratory of Pharmacology, State
Administration of Traditional Chinese Medicine, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
| | - Yingkun He
- Key Laboratory of Chinese Internal
Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
- Hebei General Hospital, Shijiazhuang,
China
| | - Pengwen Wang
- Key Laboratory of Chinese Internal
Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
- Key Laboratory of Pharmacology, State
Administration of Traditional Chinese Medicine, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
| | - Peng Wei
- Key Laboratory of Chinese Internal
Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
- Jiaozuo Hospital of Traditional Chinese
Medicine, Jiaozuo, China
| | - Huili Feng
- Key Laboratory of Chinese Internal
Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
- Key Laboratory of Pharmacology, State
Administration of Traditional Chinese Medicine, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
| | - Yingxue Rao
- Key Laboratory of Chinese Internal
Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
- University of Washington, Seattle, WA,
USA
| | - Jing Shi
- Key Laboratory of Chinese Internal
Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
- BUCM Neurology Center, Dongzhimen
Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China
| | - Jinzhou Tian
- Key Laboratory of Chinese Internal
Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing
University of Chinese Medicine (BUCM), Beijing, China
- BUCM Neurology Center, Dongzhimen
Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, China
| |
Collapse
|
84
|
Massari CM, López-Cano M, Núñez F, Fernández-Dueñas V, Tasca CI, Ciruela F. Antiparkinsonian Efficacy of Guanosine in Rodent Models of Movement Disorder. Front Pharmacol 2017; 8:700. [PMID: 29046640 PMCID: PMC5632808 DOI: 10.3389/fphar.2017.00700] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/20/2017] [Indexed: 11/18/2022] Open
Abstract
Guanosine (GUO) is a guanine-based purine nucleoside with important trophic functions and promising neuroprotective properties. Although the neuroprotective effects of GUO have been corroborated in cellular models of Parkinson’s disease (PD), its efficacy as an antiparkinsonian agent has not been fully explored in PD animal models. Accordingly, we evaluated the effectiveness of GUO in reversing motor impairments in several rodent movement disorder models, including catalepsy, tremor, and hemiparkinsonism. Our results showed that orally administered GUO antagonized reserpine-mediated catalepsy, reduced reserpine-induced tremulous jaw movements, and potentiated the number of contralateral rotations induced by L-3,4-dihydroxyphenylalanine in unilaterally 6-hydroxidopamine-lesioned rats. In addition, at 5 and 7.5 mg/kg, GUO inhibited L-DOPA-induced dyskinesia in rats chronically treated with a pro-dopaminergic agent. Overall, we describe the therapeutic potential of GUO, which may be effective not only for reversing parkinsonian motor impairments but also for reducing dyskinesia induced by treatment for PD.
Collapse
Affiliation(s)
- Caio M Massari
- Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Marc López-Cano
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, Bellvitge Institute for Biomedical Research, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Fabiana Núñez
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, Bellvitge Institute for Biomedical Research, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Víctor Fernández-Dueñas
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, Bellvitge Institute for Biomedical Research, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Carla I Tasca
- Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina, Bellvitge Institute for Biomedical Research, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
85
|
Oliveira KA, Dal-Cim TA, Lopes FG, Nedel CB, Tasca CI. Guanosine promotes cytotoxicity via adenosine receptors and induces apoptosis in temozolomide-treated A172 glioma cells. Purinergic Signal 2017; 13:305-318. [PMID: 28536931 DOI: 10.1007/s11302-017-9562-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/19/2017] [Indexed: 12/13/2022] Open
Abstract
Gliomas are a malignant tumor group whose patients have survival rates around 12 months. Among the treatments are the alkylating agents as temozolomide (TMZ), although gliomas have shown multiple resistance mechanisms for chemotherapy. Guanosine (GUO) is an endogenous nucleoside involved in extracellular signaling that presents neuroprotective effects and also shows the effect of inducing differentiation in cancer cells. The chemotherapy allied to adjuvant drugs are being suggested as a novel approach in gliomas treatment. In this way, this study evaluated whether GUO presented cytotoxic effects on human glioma cells as well as GUO effects in association with a classical chemotherapeutic compound, TMZ. Classical parameters of tumor aggressiveness, as alterations on cell viability, type of cell death, migration, and parameters of glutamatergic transmission, were evaluated. GUO (500 and 1000 μM) decreases the A172 glioma cell viability after 24, 48, or 72 h of treatment. TMZ alone or GUO plus TMZ also reduced glioma cell viability similarly. GUO combined with TMZ showed a potentiation effect of increasing apoptosis in A172 glioma cells, and a similar pattern was observed in reducing mitochondrial membrane potential. GUO per se did not elevate the acidic vesicular organelles occurrence, but TMZ or GUO plus TMZ increased this autophagy hallmark. GUO did not alter glutamate transport per se, but it prevented TMZ-induced glutamate release. GUO or TMZ did not alter glutamine synthetase activity. Pharmacological blockade of glutamate receptors did not change GUO effect on glioma viability. GUO cytotoxicity was partially prevented by adenosine receptor (A1R and A2AR) ligands. These results point to a cytotoxic effect of GUO on A172 glioma cells and suggest an anticancer effect of GUO as a putative adjuvant treatment, whose mechanism needs to be unraveled.
Collapse
Affiliation(s)
- Karen A Oliveira
- Programa de Pós-Graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Tharine A Dal-Cim
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Flávia G Lopes
- Departamento de Biologia Celular, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Cláudia B Nedel
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis, Brazil.,Departamento de Biologia Celular, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Carla Inês Tasca
- Programa de Pós-Graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, Brazil. .,Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis, Brazil. .,Departamento de Bioquímica, CCB, Universidade Federal de Santa Catarina, Trindade, Florianópolis, SC, 88040-900, Brazil.
| |
Collapse
|
86
|
The Adenosinergic System as a Therapeutic Target in the Vasculature: New Ligands and Challenges. Molecules 2017; 22:molecules22050752. [PMID: 28481238 PMCID: PMC6154114 DOI: 10.3390/molecules22050752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/24/2017] [Accepted: 05/02/2017] [Indexed: 12/20/2022] Open
Abstract
Adenosine is an adenine base purine with actions as a modulator of neurotransmission, smooth muscle contraction, and immune response in several systems of the human body, including the cardiovascular system. In the vasculature, four P1-receptors or adenosine receptors—A1, A2A, A2B and A3—have been identified. Adenosine receptors are membrane G-protein receptors that trigger their actions through several signaling pathways and present differential affinity requirements. Adenosine is an endogenous ligand whose extracellular levels can reach concentrations high enough to activate the adenosine receptors. This nucleoside is a product of enzymatic breakdown of extra and intracellular adenine nucleotides and also of S-adenosylhomocysteine. Adenosine availability is also dependent on the activity of nucleoside transporters (NTs). The interplay between NTs and adenosine receptors’ activities are debated and a particular attention is given to the paramount importance of the disruption of this interplay in vascular pathophysiology, namely in hypertension., The integration of important functional aspects of individual adenosine receptor pharmacology (such as in vasoconstriction/vasodilation) and morphological features (within the three vascular layers) in vessels will be discussed, hopefully clarifying the importance of adenosine receptors/NTs for modulating peripheral mesenteric vascular resistance. In recent years, an increase interest in purine physiology/pharmacology has led to the development of new ligands for adenosine receptors. Some of them have been patented as having promising therapeutic activities and some have been chosen to undergo on clinical trials. Increased levels of endogenous adenosine near a specific subtype can lead to its activation, constituting an indirect receptor targeting approach either by inhibition of NT or, alternatively, by increasing the activity of enzymes responsible for ATP breakdown. These findings highlight the putative role of adenosinergic players as attractive therapeutic targets for cardiovascular pathologies, namely hypertension, heart failure or stroke. Nevertheless, several aspects are still to be explored, creating new challenges to be addressed in future studies, particularly the development of strategies able to circumvent the predicted side effects of these therapies.
Collapse
|
87
|
Deng G, Qiu Z, Li D, Fang Y, Zhang S. Delayed administration of guanosine improves long‑term functional recovery and enhances neurogenesis and angiogenesis in a mouse model of photothrombotic stroke. Mol Med Rep 2017; 15:3999-4004. [PMID: 28487988 PMCID: PMC5436205 DOI: 10.3892/mmr.2017.6521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/14/2017] [Indexed: 12/20/2022] Open
Abstract
Guanosine (GUO) is neuroprotective when administered acutely for the treatment of cerebral ischemia. The aim of the present study was to investigate whether delayed administration of GUO improved long‑term functional recovery following stroke, as well as to explore the potential underlying mechanisms. GUO (8 mg/kg) or a vehicle was administered intraperitoneally for 7 consecutive days beginning 24 h prior to photothrombosis‑induced stroke in male C57/B6J mice. Behaviour tests were performed at days 1, 3, 7, 14 and 28 post‑stroke. Infarct volume was measured using Nissl staining at day 7 post‑stroke. Neurogenesis and angiogenesis were evaluated by co‑labelling bromodeoxyuridine (BrdU) with doublecortin (DCX), neuronal nuclei (NeuN) and von Willebrand factor, in immunohistochemical studies. Brain‑derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) levels in the ipsilesional brain at day 28 post‑stroke were detected by western blot analysis. Delayed administration of GUO did not reduce infarct volume or affect neurological function at day 7 post‑stroke; however, it did improve functional recovery from day 14 post‑stroke, when compared with the vehicle group. GUO significantly increased the number of BrdU+ and BrdU+/DCX+ cells in the subventricular zone and subgranular zone at all examined time points, the number of Brdu+/NeuN+ cells in the peri‑infarction region at days 14 and 28 post‑stroke and microvessel density in the peri‑infarction region at day 28 post‑stroke compared with the vehicle group. In addition, the BDNF and VEGF levels in the ipsilesional brain were significantly elevated. Delayed administration of GUO at 24 h post‑stroke enhanced neurogenesis and angiogenesis, and increased BDNF and VEGF levels, which likely contributes to long‑term functional recovery following stroke.
Collapse
Affiliation(s)
- Gang Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhandong Qiu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dayong Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yu Fang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Suming Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
88
|
Tasca C, Lanznaster D. Targeting the guanine-based purinergic system in Alzheimer's disease. Neural Regen Res 2017; 12:212-213. [PMID: 28400799 PMCID: PMC5361501 DOI: 10.4103/1673-5374.200801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
89
|
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
|
90
|
Guanosine Prevents Anhedonic-Like Behavior and Impairment in Hippocampal Glutamate Transport Following Amyloid-β1–40 Administration in Mice. Mol Neurobiol 2016; 54:5482-5496. [DOI: 10.1007/s12035-016-0082-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/23/2016] [Indexed: 12/13/2022]
|
91
|
Di Liberto V, Mudò G, Garozzo R, Frinchi M, Fernandez-Dueñas V, Di Iorio P, Ciccarelli R, Caciagli F, Condorelli DF, Ciruela F, Belluardo N. The Guanine-Based Purinergic System: The Tale of An Orphan Neuromodulation. Front Pharmacol 2016; 7:158. [PMID: 27378923 PMCID: PMC4911385 DOI: 10.3389/fphar.2016.00158] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/30/2016] [Indexed: 11/17/2022] Open
Abstract
Guanine-based purines (GBPs) have been recently proposed to be not only metabolic agents but also extracellular signaling molecules that regulate important functions in the central nervous system. In such way, GBPs-mediated neuroprotection, behavioral responses and neuronal plasticity have been broadly described in the literature. However, while a number of these functions (i.e., GBPs neurothophic effects) have been well-established, the molecular mechanisms behind these GBPs-dependent effects are still unknown. Furthermore, no plasma membrane receptors for GBPs have been described so far, thus GBPs are still considered orphan neuromodulators. Interestingly, an intricate and controversial functional interplay between GBPs effects and adenosine receptors activity has been recently described, thus triggering the hypothesis that GBPs mechanism of action might somehow involve adenosine receptors. Here, we review recent data describing the GBPs role in the brain. We focus on the involvement of GBPs regulating neuronal plasticity, and on the new hypothesis based on putative GBPs receptors. Overall, we expect to shed some light on the GBPs world since although these molecules might represent excellent candidates for certain neurological diseases management, the lack of putative GBPs receptors precludes any high throughput screening intent for the search of effective GBPs-based drugs.
Collapse
Affiliation(s)
- Valentina Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| | - Roberta Garozzo
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania Catania, Italy
| | - Monica Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| | - Víctor Fernandez-Dueñas
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Bellvitge Biomedical Research Institute, Institute of Neurosciences, University of Barcelona Barcelona, Spain
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotecnological Sciences, University of Chieti-Pescara Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotecnological Sciences, University of Chieti-Pescara Chieti, Italy
| | - Francesco Caciagli
- Department of Medical, Oral and Biotecnological Sciences, University of Chieti-Pescara Chieti, Italy
| | - Daniele F Condorelli
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania Catania, Italy
| | - Francisco Ciruela
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Bellvitge Biomedical Research Institute, Institute of Neurosciences, University of Barcelona Barcelona, Spain
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| |
Collapse
|