Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Purinergic Signal ; 15(4): 465-476, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31520282

RESUMEN

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-ß-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.


Asunto(s)
Astrocitos/efectos de los fármacos , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Guanosina/farmacología , Oxígeno/metabolismo , Animales , Astrocitos/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipoxia/metabolismo , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Purinérgicos P1/efectos de los fármacos , Receptores Purinérgicos P1/metabolismo
2.
Purinergic Signal ; 13(3): 305-318, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28536931

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Guanosina/farmacología , Receptores Purinérgicos P1/efectos de los fármacos , Antineoplásicos Alquilantes/farmacología , Autofagia/efectos de los fármacos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dacarbazina/farmacología , Glioma/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Receptores Purinérgicos P1/metabolismo , Temozolomida
3.
bioRxiv ; 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38352321

RESUMEN

Transcript buffering entails the reciprocal modulation of mRNA synthesis and degradation rates to maintain stable RNA levels under varying cellular conditions. Current research supports a global, non-sequence-specific connection between mRNA synthesis and degradation, but the underlying mechanisms are still unclear. In this study, we investigated changes in RNA metabolism following acute depletion of TIP60/KAT5, the acetyltransferase subunit of the NuA4 transcriptional coactivator complex, in mouse embryonic stem cells. By combining RNA sequencing of nuclear, cytoplasmic, and newly synthesised transcript fractions with biophysical modelling, we demonstrate that TIP60 predominantly enhances transcription of numerous genes, while a smaller set of genes undergoes TIP60-dependent transcriptional repression. Surprisingly, transcription changes caused by TIP60 depletion were offset by corresponding changes in RNA nuclear export and cytoplasmic stability, indicating gene-specific buffering mechanisms. Similarly, disruption of the unrelated ATAC coactivator complex also resulted in gene-specific transcript buffering. These findings reveal that transcript buffering functions at a gene-specific level and suggest that cells dynamically adjust RNA splicing, export, and degradation in response to individual RNA synthesis alterations, thereby sustaining cellular homeostasis.

4.
Lipids Health Dis ; 9: 142, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-21162733

RESUMEN

BACKGROUND: Statins induces cell cycle arrest, apoptosis, reduction of angiogenic factors, inhibition of the endothelial growth factor, impairing tissue adhesion and attenuation of the resistance mechanisms. The aim of this study was evaluate the anti-tumoral activity of simvastatin in a B16F10 melanoma-mouse model. METHODS: Melanoma cells were treated with different concentrations of simvastatin and assessed by viability methods. Melanoma cells (5 × 10(4)) were implanted in two month old C57Bl6/J mice. Around 7 days after cells injection, the oral treatments were started with simvastatin (5 mg/kg/day, p.o.). Tumor size, hematological and biochemical analyses were evaluated. RESULTS: Simvastatin at a concentration of 0.8 µM, 1.2 µM and 1.6 µM had toxic effect. Concentration of 1.6 µM induced a massive death in the first 24 h of incubation. Simvastatin at 0.8 µM induces early cell cycle arrest in G0/G1, followed by increase of hypodiploidy. Tumor size were evaluated and the difference of treated group and control, after ten days, demonstrates that simvastatin inhibited the tumor expansion in 68%. CONCLUSION: Simvastatin at 1.6 µM, presented cytototoxicity after 72 h of treatment, with an intense death. In vivo, simvastatin being potentially useful as an antiproliferative drug, with an impairment of growth after ten days.


Asunto(s)
Antineoplásicos/uso terapéutico , Melanoma/tratamiento farmacológico , Simvastatina/uso terapéutico , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo , Hígado/efectos de los fármacos , Masculino , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Bazo/efectos de los fármacos
5.
Math Biosci ; 324: 108346, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32371030

RESUMEN

The reaction-diffusion equation is one of the possible ways for modeling animal movement, where the reactive part stands for the population growth and the diffusive part for random dispersal of the population. However, a reaction-diffusion model may not represent all aspects of the spatial dynamics, because of the existence of distinct mechanisms that can affect the movement, such as spatial memory, which results in a bias for one direction of dispersal. This bias is modeled through an advective term on an advection-reaction-diffusion equation. Thus, considering the effects of memory on the population spread, we propose a model composed of a coupled partial differential equation system with two equations: one for the population dynamics and the other for the memory density distribution. For the population growth, we use either the exponential or logistic growth function. The analytic approach shows that for the exponential and logistic growth, the minimum traveling wave speeds are the same with or without memory dynamics in which the variation of memory is infinitesimal. From the numerical analysis, we explore how our parameters, memory, growth rate, and carrying capacity, affect the population redistribution. The combinations of these parameters result in a redistribution pattern of the population associated with either diffusive or superdiffusive and imply the dispersal is faster than the diffusion. Further, in the parameter-space defined by memory and growth rate, we have shown that memory is a factor that switches the dynamics between two spreading behaviors, one faster than the other.


Asunto(s)
Conducta Animal , Modelos Biológicos , Distribución Animal , Animales , Simulación por Computador , Ecosistema , Modelos Logísticos , Conceptos Matemáticos , Memoria , Movimiento , Dinámica Poblacional/estadística & datos numéricos , Crecimiento Demográfico
6.
Front Cell Neurosci ; 12: 376, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30459558

RESUMEN

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.

7.
Mol Neurobiol ; 55(2): 1509-1523, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28181188

RESUMEN

Malignant gliomas have resistance mechanisms to chemotherapy that enable tumor invasiveness and aggressiveness. Alternative therapies in cancer treatment, as statins, have been suggested to decrease proliferation, inhibit cell migration, and induce cell death. The aim of this study was to evaluate the effect of atorvastatin (ATOR) on cell viability, migration, proliferation, apoptosis, and autophagy in A172 human glioma cells. Temozolomide (TMZ), a chemotherapic used to glioma treatment, was tested as a comparison to cytotoxic effects on gliomas. Cell viability was also assessed in primary culture of cortical astrocytes. ATOR treatment (0.1 to 20 µM) did not alter astrocytic viability. However, in glioma cells, ATOR showed cytotoxic effect at 10 and 20 µM concentrations. TMZ (500 µM) reduced cell viability similarly to ATOR, and drug association did not show additive effect on cell viability. ATOR, TMZ, and their association decreased cell migration. ATOR also decreased glioma cell proliferation. ATOR increased apoptosis, and TMZ association showed a potentiation effect, enhancing it. ATOR and TMZ treatment increased acidic vesicular organelle (AVO) presence in A172 cells, an indicative of autophagy. ATOR effect of reducing A172 cell viability did not alter glutamate transport and glutamine synthetase activity, but it was partially prevented through antagonism of ionotropic and metabotropic glutamate receptors. Our data shows a cytotoxic effect of ATOR on glioma cells, whereas no toxicity was observed to astrocytes. ATOR showed similar cytotoxic effect as TMZ to glioma cells, and it may be a safer drug, regarding side effect induction, than chemotherapic agents.


Asunto(s)
Atorvastatina/farmacología , Neoplasias Encefálicas/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glioma/patología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Temozolomida/farmacología
8.
Neurotox Res ; 28(1): 32-42, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25827781

RESUMEN

Deposition of amyloid-ß (Aß) peptides into specific encephalic structures has been pointed as an important event related to Alzheimer's disease pathogenesis and associated with activation of glial cells, neuroinflammation, oxidative responses, and cognitive deficits. Aß-induced pro-oxidative damage may regulate the activity of glutamate transporters, leading to reduced glutamate uptake and, as a consequence, excitotoxic events. Herein, we evaluated the effects of the pretreatment of atorvastatin, a HMG-CoA reductase inhibitor, on behavioral and biochemical alterations induced by a single intracerebroventricular (i.c.v.) injection of aggregated Aß1-40 in mice. Atorvastatin (10 mg/kg/day, p.o.) was administered through seven consecutive days before Aß1-40 administration. Aß1-40 caused significant cognitive impairment in the object-place recognition task (2 weeks after the i.c.v. injection) and this phenomenon was abolished by atorvastatin pretreatment. Ex vivo evaluation of glutamate uptake into hippocampal and cerebral cortices slices showed atorvastatin, and Aß1-40 decreased hippocampal and cortical Na(+)-dependent glutamate uptake. However, Aß1-40 increased Na(+)-independent glutamate uptake and it was prevented by atorvastatin in prefrontal cortex slices. Moreover, Aß1-40 treatment significantly increased the cerebrocortical activities of glutathione reductase and glutathione peroxidase and these events were blunted by atorvastatin pretreatment. Reduced or oxidized glutathione levels were not altered by Aß1-40 and/or atorvastatin treatment. These results extend the notion of the protective action of atorvastatin against neuronal toxicity induced by Aß1-40 demonstrating that a pretreatment with atorvastatin prevents the spatial learning and memory deficits induced by Aß in rodents and promotes changes in glutamatergic and antioxidant systems mainly in prefrontal cortex.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Atorvastatina/administración & dosificación , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/prevención & control , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Fragmentos de Péptidos/toxicidad , Acetilcolinesterasa/metabolismo , Animales , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Infusiones Intraventriculares , Masculino , Ratones , Estrés Oxidativo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Aprendizaje Espacial/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA