Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Biol Chem ; 300(6): 107411, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38796067

RESUMEN

The myocyte enhancer factor (MEF2) family of transcription factors, originally discovered for its pivotal role in muscle development and function, has emerged as an essential regulator in various aspects of brain development and neuronal plasticity. The MEF2 transcription factors are known to regulate numerous important genes in the nervous system, including brain-derived neurotrophic factor (BDNF), a small secreted neurotrophin responsible for promoting the survival, growth, and differentiation of neurons. The expression of the Bdnf gene is spatiotemporally controlled by various transcription factors binding to both its proximal and distal regulatory regions. While previous studies have investigated the connection between MEF2 transcription factors and Bdnf, the endogenous function of MEF2 factors in the transcriptional regulation of Bdnf remains largely unknown. Here, we aimed to deepen the knowledge of MEF2 transcription factors and their role in the regulation of Bdnf comparatively in rat cortical and hippocampal neurons. As a result, we demonstrate that the MEF2 transcription factor-dependent enhancer located at -4.8 kb from the Bdnf gene regulates the endogenous expression of Bdnf in hippocampal neurons. In addition, we confirm neuronal activity-dependent activation of the -4.8 kb enhancer in vivo. Finally, we show that specific MEF2 family transcription factors have unique roles in the regulation of Bdnf, with the specific function varying based on the particular brain region and stimuli. Altogether, we present MEF2 family transcription factors as crucial regulators of Bdnf expression, fine-tuning Bdnf expression through both distal and proximal regulatory regions.

2.
Cell Mol Neurobiol ; 43(5): 1941-1956, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36056992

RESUMEN

Alzheimer disease (AD) is a multifactorial and age-dependent neurodegenerative disorder, whose pathogenesis, classically associated with the formation of senile plaques and neurofibrillary tangles, is also dependent on oxidative stress and neuroinflammation chronicization. Currently, the standard symptomatic therapy, based on acetylcholinesterase inhibitors, showed a limited therapeutic potential, whereas disease-modifying treatment strategies are still under extensive research. Previous studies have demonstrated that Oxotremorine-M (Oxo), a non-selective muscarinic acetylcholine receptors agonist, exerts neurotrophic functions in primary neurons, and modulates oxidative stress and neuroinflammation phenomena in rat brain. In the light of these findings, in this study, we aimed to investigate the neuroprotective effects of Oxo treatment in an in vitro model of AD, represented by differentiated SH-SY5Y neuroblastoma cells exposed to Aß1-42 peptide. The results demonstrated that Oxo treatment enhances cell survival, increases neurite length, and counteracts DNA fragmentation induced by Aß1-42 peptide. The same treatment was also able to block oxidative stress and mitochondria morphological/functional impairment associated with Aß1-42 cell exposure. Overall, these results suggest that Oxo, by modulating cholinergic neurotransmission, survival, oxidative stress response, and mitochondria functionality, may represent a novel multi-target drug able to achieve a therapeutic synergy in AD. Illustration of the main pathological hallmarks and mechanisms underlying AD pathogenesis, including neurodegeneration and oxidative stress, efficiently counteracted by treatment with Oxo, which may represent a promising therapeutic molecule. Created with BioRender.com under academic license.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Ratas , Animales , Humanos , Antioxidantes/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Oxotremorina/farmacología , Enfermedades Neuroinflamatorias , Acetilcolinesterasa , Péptidos beta-Amiloides , Neuroblastoma/patología , Receptores Muscarínicos
3.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-35163472

RESUMEN

Over the last decade, several compounds have been identified for the treatment of obesity. However, due to the complexity of the disease, many pharmacological interventions have raised concerns about their efficacy and safety. Therefore, it is important to discover new factors involved in the induction/progression of obesity. Adipose stromal/stem cells (ASCs), which are mostly isolated from subcutaneous adipose tissue, are the primary cells contributing to the expansion of fat mass. Like other cells, ASCs release nanoparticles known as extracellular vesicles (EVs), which are being actively studied for their potential applications in a variety of diseases. Here, we focused on the importance of the con-tribution of ASC-derived EVs in the regulation of metabolic processes. In addition, we outlined the advantages/disadvantages of the use of EVs as potential next-generation anti-obesity agents.


Asunto(s)
Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/citología , Obesidad/metabolismo , Adipogénesis , Vesículas Extracelulares/trasplante , Homeostasis , Humanos , Obesidad/terapia , Grasa Subcutánea/citología , Grasa Subcutánea/metabolismo
4.
Int J Mol Sci ; 23(16)2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-36012187

RESUMEN

The low-density-lipoprotein receptors represent a family of pleiotropic cell surface receptors involved in lipid homeostasis, cell migration, proliferation and differentiation. The family shares common structural features but also has significant differences mainly due to tissue-specific interactors and to peculiar proteolytic processing. Among the receptors in the family, recent studies place low-density lipoprotein receptor-related protein 8 (LRP8) at the center of both neurodegenerative and cancer-related pathways. From one side, its overexpression has been highlighted in many types of cancer including breast, gastric, prostate, lung and melanoma; from the other side, LRP8 has a potential role in neurodegeneration as apolipoprotein E (ApoE) and reelin receptor, which are, respectively, the major risk factor for developing Alzheimer's disease (AD) and the main driver of neuronal migration, and as a γ-secretase substrate, the main enzyme responsible for amyloid formation in AD. The present review analyzes the contributions of LDL receptors, specifically of LRP8, in both cancer and neurodegeneration, pointing out that depending on various interactions and peculiar processing, the receptor can contribute to both proliferative and neurodegenerative processes.


Asunto(s)
Enfermedad de Alzheimer , Neoplasias , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Humanos , Lipoproteínas LDL , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Receptores de LDL/metabolismo
5.
Int J Mol Sci ; 23(15)2022 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-35955821

RESUMEN

Recent studies have highlighted the mechanisms controlling the formation of cerebral cholesterol, which is synthesized in situ primarily by astrocytes, where it is loaded onto apolipoproteins and delivered to neurons and oligodendrocytes through interactions with specific lipoprotein receptors. The "cholesterol shuttle" is influenced by numerous proteins or carbohydrates, which mainly modulate the lipoprotein receptor activity, function and signaling. These molecules, provided with enzymatic/proteolytic activity leading to the formation of peptide fragments of different sizes and specific sequences, could be also responsible for machinery malfunctions, which are associated with neurological, neurodegenerative and neurodevelopmental disorders. In this context, we have pointed out that purines, ancestral molecules acting as signal molecules and neuromodulators at the central nervous system, can influence the homeostatic machinery of the cerebral cholesterol turnover and vice versa. Evidence gathered so far indicates that purine receptors, mainly the subtypes P2Y2, P2X7 and A2A, are involved in the pathogenesis of neurodegenerative diseases, such as Alzheimer's and Niemann-Pick C diseases, by controlling the brain cholesterol homeostasis; in addition, alterations in cholesterol turnover can hinder the purine receptor function. Although the precise mechanisms of these interactions are currently poorly understood, the results here collected on cholesterol-purine reciprocal control could hopefully promote further research.


Asunto(s)
Sistema Nervioso Central , Enfermedades de Niemann-Pick , Sistema Nervioso Central/metabolismo , Colesterol/metabolismo , Humanos , Neuronas/metabolismo , Enfermedades de Niemann-Pick/metabolismo , Purinas/metabolismo , Receptores Purinérgicos/metabolismo
6.
Int J Mol Sci ; 24(1)2022 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-36613467

RESUMEN

Astrocytic networks and gap junctional communication mediated by connexins (Cxs) have been repeatedly implicated in seizures, epileptogenesis, and epilepsy. However, the effect of seizures on Cx expression is controversial. The present study focused on the response of Cxs to status epilepticus (SE), which is in turn an epileptogenic insult. The expression of neuronal Cx36 and astrocytic Cx30 and Cx43 mRNAs was investigated in the brain of rats in the first day after pilocarpine-induced SE. In situ hybridization revealed a progressive decrease in Cx43 and Cx30 mRNA levels, significantly marked 24 h after SE onset in neocortical areas and the hippocampus, and in most thalamic domains, whereas Cx36 mRNA did not exhibit obvious changes. Regional evaluation with quantitative real-time-RT-PCR confirmed Cx43 and Cx30 mRNA downregulation 24 h after SE, when ongoing neuronal cell death was found in the same brain regions. Immunolabeling showed at the same time point marked a decrease in Cx43, microglia activation, and interleukin-1ß induction in some microglial cells. The data showed a transient downregulation of astroglial Cxs in the cortical and thalamic areas in which SE triggers neurodegenerative events in concomitance with microglia activation and cytokine expression. This could potentially represent a protective response of neuroglial networks to SE-induced acute damage.


Asunto(s)
Conexinas , Estado Epiléptico , Animales , Ratas , Astrocitos/metabolismo , Conexina 43/metabolismo , Conexinas/metabolismo , Regulación hacia Abajo , Hipocampo/metabolismo , Pilocarpina/toxicidad , ARN Mensajero/metabolismo , Convulsiones/metabolismo , Estado Epiléptico/inducido químicamente , Estado Epiléptico/genética , Estado Epiléptico/metabolismo
7.
Int J Mol Sci ; 21(23)2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-33291390

RESUMEN

Acute or chronic administration of guanosine (GUO) induces anxiolytic-like effects, for which the adenosine (ADO) system involvement has been postulated yet without a direct experimental evidence. Thus, we aimed to investigate whether adenosine receptors (ARs) are involved in the GUO-mediated anxiolytic-like effect, evaluated by three anxiety-related paradigms in rats. First, we confirmed that acute treatment with GUO exerts an anxiolytic-like effect. Subsequently, we investigated the effects of pretreatment with ADO or A1R (CPA, CCPA) or A2AR (CGS21680) agonists 10 min prior to GUO on a GUO-induced anxiolytic-like effect. All the combined treatments blocked the GUO anxiolytic-like effect, whereas when administered alone, each compound was ineffective as compared to the control group. Interestingly, the pretreatment with nonselective antagonist caffeine or selective A1R (DPCPX) or A2AR (ZM241385) antagonists did not modify the GUO-induced anxiolytic-like effect. Finally, binding assay performed in hippocampal membranes showed that [3H]GUO binding became saturable at 100-300 nM, suggesting the existence of a putative GUO binding site. In competition experiments, ADO showed a potency order similar to GUO in displacing [3H]GUO binding, whereas AR selective agonists, CPA and CGS21680, partially displaced [3H]GUO binding, but the sum of the two effects was able to displace [3H]GUO binding to the same extent of ADO alone. Overall, our results strengthen previous data supporting GUO-mediated anxiolytic-like effects, add new evidence that these effects are blocked by A1R and A2AR agonists and pave, although they do not elucidate the mechanism of GUO and ADO receptor interaction, for a better characterization of GUO binding sites in ARs.


Asunto(s)
Ansiedad/etiología , Ansiedad/metabolismo , Guanosina/efectos adversos , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Animales , Ansiedad/psicología , Conducta Animal , Membrana Celular/metabolismo , Oscuridad , Relación Dosis-Respuesta a Droga , Guanosina/metabolismo , Hipocampo/metabolismo , Luz , Ratas , Receptor de Adenosina A1/genética , Receptor de Adenosina A2A/genética
8.
J Neuroinflammation ; 16(1): 44, 2019 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-30777084

RESUMEN

BACKGROUND: Aß1-42 peptide abnormal production is associated with the development and maintenance of neuroinflammation and oxidative stress in brains from Alzheimer disease (AD) patients. Suppression of neuroinflammation may then represent a suitable therapeutic target in AD. We evaluated the efficacy of IFNß1a in attenuating cognitive impairment and inflammation in an animal model of AD. METHODS: A rat model of AD was obtained by intra-hippocampal injection of Aß1-42 peptide (23 µg/2 µl). After 6 days, 3.6 µg of IFNß1a was given subcutaneously (s.c.) for 12 days. Using the novel object recognition (NOR) test, we evaluated changes in cognitive function. Measurement of pro-inflammatory or anti-inflammatory cytokines, reactive oxygen species (ROS), and SOD activity levels was performed in the hippocampus. Data were evaluated by one-way ANOVA with Fisher's Protected Least Significant Difference (PLSD) test. RESULTS: We showed that treatment with IFNß1a was able to reverse memory impairment and to counteract microglia activation and upregulation of pro-inflammatory cytokines (IL-6, IL-1ß) in the hippocampus of Aß1-42-injected rats. The anti-inflammatory cytokine IL-10, significantly reduced in the Aß1-42 animals, recovered to control levels following IFNß1a treatment. IFNß1a also reduced ROS and lipids peroxidation and increased SOD1 protein levels in the hippocampus of Aß1-42-injected rats. CONCLUSION: This study shows that IFNß1a is able to reverse the inflammatory and cognitive effects of intra-hippocampal Aß1-42 in the rat. Given the role played by inflammation in AD pathogenesis and the established efficacy of IFNß1a in the treatment of inflammatory diseases of the central nervous system such as multiple sclerosis, its use may be a viable strategy to inhibit the pro-inflammatory cytokine and oxidative stress cascade associated with Aß deposition in the hippocampus of AD patients.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Inflamación/tratamiento farmacológico , Inflamación/etiología , Interferón beta-1a/uso terapéutico , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/toxicidad , Animales , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Peroxidación de Lípido/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Fragmentos de Péptidos/toxicidad , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/metabolismo , Factores de Tiempo
9.
J Cell Physiol ; 233(8): 6107-6116, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29323700

RESUMEN

The cholinergic system plays a crucial role in modulating in the central nervous system physiological responses such as neurogenesis, neuronal differentiation, synaptic plasticity, and neuroprotection. In a recent study, we showed that Oxotremorine-M, a non-selective muscarinic acetylcholine receptor agonist, is able to transactivate the fibroblast growth factor receptor and to produce a significant increase in the hippocampal primary neurite outgrowth. In the present study we aimed to explore in the rat hippocampus the possible effect of acute or chronic treatment with Oxotremorine-M on some heat shock proteins (Hsp60, Hsp70, Hsp90) and on activation of related transcription factor heat shock factor 1 (HSF1). Following single injection of Oxotremorine-M (0.4 mg/kg) all Hsps examined were significantly increased in at least one of the time points studied (24, 48, and 72 hr). Treatment with Oxotremorine-M significantly increased the level of phosphorylated HSF1 in all time points studied, without change of protein levels. Similar pattern of Hsps changes was obtained following chronic Oxotremorine-M treatment (0.2 mg/kg) for 5 days. Surprisingly, following chronic treatment for 10 days no changes were observed in Hsps. The muscarinic acetylcholine receptor antagonist scopolamine (1 mg/kg) was able to completely block Oxotremorine-M effects on Hsps. In conclusion, considering the function of Hsps in protecting neuronal cells from deleterious proteotoxic stress, for example, protein mis-folding and aggregation, the results obtained indicate that muscarinic acetylcholine receptor activation may have implications in potential treatment of neurodegenerative disorders linked to protein aggregation, such as Alzheimer disease.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Hipocampo/metabolismo , Receptores Muscarínicos/metabolismo , Animales , Factores de Transcripción del Choque Térmico/metabolismo , Hipocampo/efectos de los fármacos , Masculino , Enfermedades Neurodegenerativas/metabolismo , Proyección Neuronal/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxotremorina/análogos & derivados , Oxotremorina/farmacología , Ratas , Ratas Wistar , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Escopolamina/farmacología , Transducción de Señal/efectos de los fármacos
10.
J Cell Physiol ; 232(8): 2044-2052, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27576008

RESUMEN

In the mdx mice model of Duchenne Muscular Dystrophy (DMD), mild endurance exercise training positively affected limb skeletal muscles, whereas few and controversial data exist on the effects of training on the diaphragm. The diaphragm was examined in mdx (C57BL/10ScSn-Dmdmdx) and wild-type (WT, C57BL/10ScSc) mice under sedentary conditions (mdx-SD, WT-SD) and during mild exercise training (mdx-EX, WT-EX). At baseline, and after 30 and 45 days (training: 5 d/wk for 6 weeks), diaphragm muscle morphology and Cx39 protein were assessed. In addition, tissue levels of the chaperonins Hsp60 and Hsp70 and the p65 subunit of nuclear factor-kB (NF-kB) were measured in diaphragm, gastrocnemius, and quadriceps in each experimental group at all time points. Although morphological analysis showed unchanged total area of necrosis/regeneration in the diaphragm after training, there was a trend for larger areas of regeneration than necrosis in the diaphragm of mdx-EX compared to mdx-SD mice. However, the levels of Cx39, a protein associated with active regeneration in damaged muscle, were similar in the diaphragm of mdx-EX and mdx-SD mice. Hsp60 significantly decreased at 45 days in the diaphragm, but not in limb muscles, in both trained and sedentary mdx compared to WT mice. In limb muscles, but not in the diaphragm, Hsp70 and NF-kB p65 levels were increased in mdx mice irrespective of training at 30 and 45 days. Therefore, the diaphragm of mdx mice showed little inflammatory and stress responses over time, and appeared hardly affected by mild endurance training. J. Cell. Physiol. 232: 2044-2052, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Diafragma/fisiopatología , Terapia por Ejercicio/métodos , Fuerza Muscular , Distrofia Muscular de Duchenne/terapia , Animales , Chaperonina 60/metabolismo , Conexinas/metabolismo , Diafragma/metabolismo , Diafragma/patología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Proteínas HSP70 de Choque Térmico/metabolismo , Masculino , Ratones Endogámicos mdx , Proteínas Mitocondriales/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Necrosis , Fenotipo , Resistencia Física , Factores de Tiempo , Factor de Transcripción ReIA/metabolismo
11.
Cells ; 13(4)2024 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-38391968

RESUMEN

In this study, we investigated the beneficial effects of grapefruit IntegroPectin, derived from industrial waste grapefruit peels via hydrodynamic cavitation, on microglia cells exposed to oxidative stress conditions. Grapefruit IntegroPectin fully counteracted cell death and the apoptotic process induced by cell exposure to tert-butyl hydroperoxide (TBH), a powerful hydroperoxide. The protective effects of the grapefruit IntegroPectin were accompanied with a decrease in the amount of ROS, and were strictly dependent on the activation of the phosphoinositide 3-kinase (PI3K)/Akt cascade. Finally, IntegroPectin treatment inhibited the neuroinflammatory response and the basal microglia activation by down-regulating the PI3K- nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB)- inducible nitric oxide synthase (iNOS) cascade. These data strongly support further investigations aimed at exploring IntegroPectin's therapeutic role in in vivo models of neurodegenerative disorders, characterized by a combination of chronic neurodegeneration, oxidative stress and neuroinflammation.


Asunto(s)
Citrus paradisi , Microglía , Humanos , Microglía/metabolismo , Citrus paradisi/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/metabolismo , Línea Celular
12.
Chemosphere ; 359: 142278, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38734249

RESUMEN

Different bioactive molecules extracted from macroalgae, including oxylipins, showed interesting potentials in different applications, from healthcare to biomaterial manufacturing and environmental remediation. Thus far, no studies reported the effects of oxylipins-containing macroalgae extracts on embryo development of marine invertebrates and on neuroblastoma cancer cells. Here, the effects of an oxylipins-containing extract from Ericaria brachycarpa, a canopy-forming brown algae, were investigated on the development of Arbacia lixula sea urchin embryos and on SH-SY5Y neuroblastoma cells viability. Embryos and cells were exposed to concentrations covering a full 0-100% dose-response curve, with doses ranging from 0 to 40 µg mL-1 for embryos and from 0 to 200 µg mL-1 for cells. These natural marine toxins caused a dose-dependent decrease of normal embryos development and of neuroblastoma cells viability. Toxicity was higher for exposures starting from the gastrula embryonal stage if compared to the zygote and pluteus stages, with an EC50 significantly lower by 33 and 68%, respectively. Embryos exposed to low doses showed a general delay in development with a decrease in the ability to calcify, while higher doses caused 100% block of embryo growth. Exposure of SH-SY5Y neuroblastoma cells to 40 µg mL-1 for 72 h caused 78% mortality, while no effect was observed on their neuronal-like cells derivatives, suggesting a selective targeting of proliferating cells. Western Blot experiments on both model systems displayed the modulation of different molecular markers (HSP60, HSP90, LC3, p62, CHOP and cleaved caspase-7), showing altered stress response and enhanced autophagy and apoptosis, confirmed by increased fragmented DNA in apoptotic nuclei. Our study gives new insights into the molecular strategies that marine invertebrates use when responding to their environmental natural toxins and suggests the E. brachycarpa's extract as a potential source for the development of innovative, environmentally friendly products with larvicide and antineoplastic activity.


Asunto(s)
Supervivencia Celular , Neuroblastoma , Oxilipinas , Erizos de Mar , Animales , Supervivencia Celular/efectos de los fármacos , Erizos de Mar/efectos de los fármacos , Humanos , Oxilipinas/farmacología , Línea Celular Tumoral , Algas Marinas , Apoptosis/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Phaeophyceae/química , Desarrollo Embrionario/efectos de los fármacos , Toxinas Marinas/toxicidad
13.
Antioxidants (Basel) ; 12(3)2023 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-36978935

RESUMEN

The differentiation of neural progenitors is a complex process that integrates different signals to drive transcriptional changes, which mediate metabolic, electrophysiological, and morphological cellular specializations. Understanding these adjustments is essential within the framework of stem cell and cancer research and therapy. Human neuroblastoma SH-SY5Y cells, widely used in neurobiology research, can be differentiated into neuronal-like cells through serum deprivation and retinoic acid (RA) supplementation. In our study, we observed that the differentiation process triggers the expression of Heat Shock Protein 70 (HSP70). Notably, inhibition of HSP70 expression by KNK437 causes a dramatic increase in cell death. While undifferentiated SH-SY5Y cells show a dose-dependent decrease in cell survival following exposure to hydrogen peroxide (H2O2), differentiated cells become resistant to H2O2-induced cell death. Interestingly, the differentiation process enhances the expression of SOD1 protein, and inhibition of HSP70 expression counteracts this effect and increases the susceptibility of differentiated cells to H2O2-induced cell death, suggesting that the cascade HSP70-SOD1 is involved in promoting survival against oxidative stress-dependent damage. Treatment of differentiated SH-SY5Y cells with Oxotremorine-M (Oxo), a muscarinic acetylcholine receptor agonist, enhances the expression of HSP70 and SOD1 and counteracts tert-Butyl hydroperoxide-induced cell death and reactive oxygen species (ROS) generation. It is worth noting that co-treatment with KNK437 reduces SOD1 expression and Oxo-induced protection against oxidative stress damage, suggesting the involvement of HSP70/SOD1 signaling in this beneficial effect. In conclusion, our findings demonstrate that manipulation of the HSP70 signal modulates SH-SY5Y differentiation and susceptibility to oxidative stress-dependent cell death and unravels novel mechanisms involved in Oxo neuroprotective functions. Altogether these data provide novel insights into the mechanisms underlying neuronal differentiation and preservation under stress conditions.

14.
Antioxidants (Basel) ; 10(4)2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-33916762

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked recessive progressive lethal disorder caused by the lack of dystrophin, which determines myofibers mechanical instability, oxidative stress, inflammation, and susceptibility to contraction-induced injuries. Unfortunately, at present, there is no efficient therapy for DMD. Beyond several promising gene- and stem cells-based strategies under investigation, physical activity may represent a valid noninvasive therapeutic approach to slow down the progression of the pathology. However, ethical issues, the limited number of studies in humans and the lack of consistency of the investigated training interventions generate loss of consensus regarding their efficacy, leaving exercise prescription still questionable. By an accurate analysis of data about the effects of different protocol of exercise on muscles of mdx mice, the most widely-used pre-clinical model for DMD research, we found that low intensity exercise, especially in the form of low speed treadmill running, likely represents the most suitable exercise modality associated to beneficial effects on mdx muscle. This protocol of training reduces muscle oxidative stress, inflammation, and fibrosis process, and enhances muscle functionality, muscle regeneration, and hypertrophy. These conclusions can guide the design of appropriate studies on human, thereby providing new insights to translational therapeutic application of exercise to DMD patients.

15.
Front Pharmacol ; 12: 658806, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33986683

RESUMEN

Neuroblastoma arises from neural crest cell precursors failing to complete the process of differentiation. Thus, agents helping tumor cells to differentiate into normal cells can represent a valid therapeutic strategy. Here, we evaluated whether guanosine (GUO), a natural purine nucleoside, which is able to induce differentiation of many cell types, may cause the differentiation of human neuroblastoma SH-SY5Y cells and the molecular mechanisms involved. We found that GUO, added to the cell culture medium, promoted neuron-like cell differentiation in a time- and concentration-dependent manner. This effect was mainly due to an extracellular GUO action since nucleoside transporter inhibitors reduced but not abolished it. Importantly, GUO-mediated neuron-like cell differentiation was independent of adenosine receptor activation as it was not altered by the blockade of these receptors. Noteworthy, the neuritogenic activity of GUO was not affected by blocking the phosphoinositide 3-kinase pathway, while it was reduced by inhibitors of protein kinase C or soluble guanylate cyclase. Furthermore, the inhibitor of the enzyme heme oxygenase-1 but not that of nitric oxide synthase reduced GUO-induced neurite outgrowth. Interestingly, we found that GUO was largely metabolized into guanine by the purine nucleoside phosphorylase (PNP) enzyme released from cells. Taken together, our results suggest that GUO, promoting neuroblastoma cell differentiation, may represent a potential therapeutic agent; however, due to its spontaneous extracellular metabolism, the role played by the GUO-PNP-guanine system needs to be further investigated.

16.
Sci Rep ; 9(1): 14233, 2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31578381

RESUMEN

Recently we found that acute treatment with Oxotremorine (Oxo), a non-selective mAChRs agonist, up-regulates heat shock proteins and activates their transcription factor heat shock factor 1 in the rat hippocampus. Here we aimed to investigate: a) if acute treatment with Oxo may regulate pro-inflammatory or anti-inflammatory cytokines and oxidative stress in the rat hippocampus; b) if chronic restraint stress (CRS) induces inflammatory or oxidative alterations in the hippocampus and whether such alterations may be affected by chronic treatment with Oxo. In the acute experiment, rats were injected with single dose of Oxo (0.4 mg/kg) and sacrificed at 24 h, 48 h and 72 h. In the CRS experiment, the rats were exposed for 21 days to the CRS and then were treated with Oxo (0.2 mg/kg) for further 10 days. The acute Oxo treatment showed an ability to significantly reduce reactive oxygen species (ROS), singlet oxygen (1O2), pro-inflammatory cytokines levels (IL-1ß and IL-6) and phosphorylated NF-κB-p65. Acute Oxo treatment also increased superoxide dismutase (SOD)-2 protein levels and stimulated SOD activity. No differences were detected in the anti-inflammatory cytokine levels, including IL-10 and TGF-ß1. In the group of rats exposed to the CRS were found increased hippocampal IL-1ß and IL-6 levels, together with a reduction of SOD activity level. These changes produced by CRS were counteracted by chronic Oxo treatment. In contrast, the upregulation of ROS and 1O2 levels in the CRS group was not counteracted by chronic Oxo treatment. The results revealed a hippocampal anti-inflammatory and antioxidant effect of Oxo treatment in both basal conditions and anti-inflammatory in the CRS rat model.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Hipocampo/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Fármacos Neuroprotectores/farmacología , Oxotremorina/farmacología , Receptores Muscarínicos/efectos de los fármacos , Animales , Hipocampo/metabolismo , Hidrocortisona/sangre , Inflamación , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-6/biosíntesis , Interleucina-6/genética , Masculino , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Receptores Muscarínicos/metabolismo , Restricción Física/efectos adversos , Escopolamina/farmacología , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Superóxido Dismutasa-1/biosíntesis , Superóxido Dismutasa-1/genética , Factor de Transcripción ReIA/metabolismo
17.
Brain Res ; 1193: 12-24, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18190895

RESUMEN

Precursor cell proliferation is present in the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus of the hippocampus of adult rat and persists during aging although at reduced levels. Previous studies have shown that acute intermittent nicotine treatment significantly increases fibroblast growth factor-2 (FGF-2) expression in several brain regions of aged rats. The aim of the present investigation was to test the hypothesis that nicotine-induced expression of FGF-2 may restore the age-related decline of precursor cell proliferation. It was first demonstrated that nicotine treatment increases both mRNA and protein FGF-2 in the SVZ of aged male rats (18 months old). The effect of nicotine on precursor cell proliferation in the SVZ was studied by i.p. injection of 5-bromo-2'-deoxyuridine (BrdU) 40 mg/kg to label dividing cells. The nicotine treatment was found to significantly enhance precursor cell proliferation in the SVZ. This increase was sufficiently large to restore the age-related decline of proliferating precursor cells observed in aged rats to that found in young adult rats (3 months old). FGF-2 was expressed in GFAP-positive cells and may act via its receptor FGFR1 that was found expressed in nestin-positive cells of the SVZ. The data obtained demonstrated that the age-related decline of precursor cell proliferation may be counteracted by activating a trophic mechanism mediated by FGF-2.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Envejecimiento , Proliferación Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Ventrículos Laterales/citología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Análisis de Varianza , Animales , Anticuerpos/farmacología , Encéfalo/anatomía & histología , Bromodesoxiuridina/metabolismo , Recuento de Células , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Nestina , Ratas , Ratas Wistar
18.
Neurosci Lett ; 447(1): 20-5, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18835325

RESUMEN

Several findings have suggested the existence in the subventricular zone (SVZ) from sagittal sections of adult rat brain of a trophic mechanism, mediated by fibroblast growth factor-2 (FGF-2) and its multiple high-affinity FGF receptors (FGFRs), regulating neurogenesis mainly by controlling precursor cell proliferation. However, no clear data are available on the expression of FGF-2 and FGFRs in proliferating precursor cells of the SVZ. To address these questions we examined FGF-2 mRNA and its FGFR mRNA expression in proliferating precursor cells of the SVZ by using a double labeling procedure, combining in situ hybridization for FGF-2 and its FGFR mRNA with immunohistochemistry for bromodeoxyuridine (BrdU), a marker for proliferating cells. The results showed that FGFR1 and FGFR2 mRNAs were expressed in BrdU-labeled proliferating precursor cells, whereas FGFR3 and FGF-2 mRNAs were not, suggesting that in the SVZ the proliferating precursor cells express FGFR1 or FGFR2 and they may respond to FGF-2 released by non-proliferating cells. The FGFR4 mRNA was not found expressed in the SVZ. In the future, by identifying the cell types expressing FGFRs, it will be possible to gain insight into the functional activity of FGF2 within the SVZ.


Asunto(s)
Células Madre Adultas/metabolismo , Proliferación Celular , Ventrículos Cerebrales/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Encéfalo/anatomía & histología , Bromodesoxiuridina/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Expresión Génica/fisiología , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo
19.
Front Pharmacol ; 9: 110, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29515443

RESUMEN

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.

20.
Auton Neurosci ; 210: 55-64, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29305058

RESUMEN

Mutations in the HGPRT1 gene, which encodes hypoxanthine-guanine phosphoribosyltransferase (HGprt), housekeeping enzyme responsible for recycling purines, lead to Lesch-Nyhan disease (LND). Clinical expression of LND indicates that HGprt deficiency has adverse effects on gastrointestinal motility. Therefore, we aimed to evaluate intestinal motility in HGprt knockout mice (HGprt¯). Spontaneous and neurally evoked mechanical activity was recorded in vitro as changes in isometric tension in circular muscle strips of distal colon. HGprt¯ tissues showed a lower in amplitude spontaneous activity and atropine-sensitivity neural contraction compared to control mice. The responses to carbachol and to high KCl were reduced, demonstrating a widespread impairment of contractility. L-NAME was not able in the HGprt¯ tissues to restore the large amplitude contractile activity typical of control. In HGprt¯ colon, a reduced expression of dopaminergic D1 receptor was observed together with the loss of its tonic inhibitory activity present in control-mice. The analysis of inflammatory and oxidative stress in colonic tissue of HGprt¯ mice revealed a significant increase of lipid peroxidation associated with over production of oxygen free radicals. In conclusion, HGprt deficiency in mice is associated with a decrease in colon contractility, not dependent upon reduction of acetylcholine release from the myenteric plexus or hyperactivity of inhibitory signalling. By contrast the increased levels of oxidative stress could partially explain the reduced colon motility in HGprt¯ mice. Colonic dysmotility observed in HGprt¯ mice may mimic the gastrointestinal dysfunctions symptoms of human syndrome, providing a useful animal model to elucidate the pathophysiology of this problem in the LND.


Asunto(s)
Motilidad Gastrointestinal/genética , Regulación de la Expresión Génica/genética , Síndrome de Lesch-Nyhan/complicaciones , Músculo Liso/fisiopatología , Animales , Atropina/farmacología , Benzazepinas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Carbacol/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Inhibidores Enzimáticos/farmacología , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/genética , Cara , Motilidad Gastrointestinal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipoxantina Fosforribosiltransferasa/deficiencia , Hipoxantina Fosforribosiltransferasa/metabolismo , Técnicas In Vitro , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/patología , Síndrome de Lesch-Nyhan/fisiopatología , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Neurotransmisores/farmacología , Especies Reactivas de Oxígeno/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA