Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Am J Transplant ; 19(12): 3240-3249, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31152486

RESUMEN

The mechanisms of tacrolimus-induced ß cell toxicity are unknown. Tacrolimus (TAC) and rapamycin (Rapa) both bind to FK506-binding protein 12 (FKBP12). Also, both molecular structures are similar. Because of this similarity, we hypothesized that TAC can also inhibit the mTOR signalling, constituting a possible mechanism of ß cell toxicity. Thus, we studied the effect of TAC and Rapa over the mTOR pathway, v-maf musculoaponeurotic fibrosarcoma oncogene homolog A (MafA), and insulin secretion and content in INS-1 ß cells treated with or without glucose and palmitate and in islets from lean or obese rats. TAC and Rapa inhibited the mTOR pathway as reflected by lower levels of phospho-mTOR, phospo-p70S6K, and phospo-S6. The effect of Rapa was larger than TAC. Both drugs reduced the levels of MafA, insulin secretion, and content although these effects were larger with TAC. The changes on MafA and insulin metabolism were observed in cells on glucose and palmitate, in obese animals, and were absent in cells on maintenance medium or in lean animals. In silico docking and immunoprecipitation experiments confirmed that TAC can form a stable noncovalent interaction with FKBP12-mTOR. Thus, the mTOR inhibition may be a mechanism contributing to the diabetogenic effect of TAC.


Asunto(s)
Apoptosis , Diabetes Mellitus Experimental/patología , Células Secretoras de Insulina/patología , Obesidad/fisiopatología , Serina-Treonina Quinasas TOR/metabolismo , Tacrolimus/toxicidad , Delgadez/fisiopatología , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Inmunosupresores/toxicidad , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratas , Ratas Zucker , Transducción de Señal
2.
J Neurogenet ; 31(4): 266-287, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28762842

RESUMEN

Eating-disorders (EDs) consequences to human health are devastating, involving social, mental, emotional, physical and life-threatening aspects, concluding on impairment and death in cases of extreme anorexia nervosa. It also implies that people suffering an ED need to find psychiatric and psychological help as soon as possible to achieve a fully physical and emotional recovery. Unfortunately, to date, there is a crucial lack of efficient clinical treatment to these disorders. In this review, we present an overview concerning the actual pharmacological and psychological treatments, the knowledge of cells, circuits, neuropeptides, neuromodulators and hormones in the human brain- and other organs- underlying these disorders, the studies in animal models and, finally, the genetic approaches devoted to face this challenge. We will also discuss the need for new perspectives, avenues and strategies to be developed in order to pave the way to novel and more efficient therapeutics.


Asunto(s)
Anorexia Nerviosa/genética , Trastorno por Atracón/genética , Bulimia Nerviosa/genética , Anorexia Nerviosa/metabolismo , Trastorno por Atracón/metabolismo , Bulimia Nerviosa/metabolismo , Predisposición Genética a la Enfermedad , Humanos
3.
Development ; 139(17): 3200-10, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22872087

RESUMEN

The establishment of neural circuits depends on the ability of axonal growth cones to sense their surrounding environment en route to their target. To achieve this, a coordinated rearrangement of cytoskeleton in response to extracellular cues is essential. Although previous studies have identified different chemotropic and adhesion molecules that influence axonal development, the molecular mechanism by which these signals control the cytoskeleton remains poorly understood. Here, we show that in vivo conditional ablation of the focal adhesion kinase gene (Fak) from mouse hippocampal pyramidal cells impairs axon outgrowth and growth cone morphology during development, which leads to functional defects in neuronal connectivity. Time-lapse recordings and in vitro FRAP analysis indicate that filopodia motility is altered in growth cones lacking FAK, probably owing to deficient actin turnover. We reveal the intracellular pathway that underlies this process and describe how phosphorylation of the actin nucleation-promoting factor N-WASP is required for FAK-dependent filopodia formation. Our study reveals a novel mechanism through which FAK controls filopodia formation and actin nucleation during axonal development.


Asunto(s)
Actinas/metabolismo , Axones/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Red Nerviosa/crecimiento & desarrollo , Neuronas/citología , Seudópodos/fisiología , Animales , Axones/enzimología , Cartilla de ADN/genética , Electroporación , Potenciales Postsinápticos Excitadores/fisiología , Recuperación de Fluorescencia tras Fotoblanqueo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Conos de Crecimiento/fisiología , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Transgénicos , Faloidina , Seudópodos/enzimología
4.
J Neurosci ; 31(8): 2721-33, 2011 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-21414895

RESUMEN

The possibility of changing the number of synapses may be an important asset in the treatment of neurological diseases. In this context, the synaptogenic role of the phosphoinositide-3-kinase (PI3K) signaling cascade has been previously demonstrated in Drosophila. This study shows that treatment with a PI3K-activating transduction peptide is able to promote synaptogenesis and spinogenesis in primary cultures of rat hippocampal neurons, as well as in CA1 hippocampal neurons in vivo. In culture, the peptide increases synapse density independently of cell density, culture age, dendritic complexity, or synapse type. The induced synapses also increase neurotransmitter release from cultured neurons. The synaptogenic signaling pathway includes PI3K-Akt. Furthermore, the treatment is effective on adult neurons, where it induces spinogenesis and enhances the cognitive behavior of treated animals in a fear-conditioning assay. These findings demonstrate that functional synaptogenesis can be induced in mature mammalian brains through PI3K activation.


Asunto(s)
Región CA1 Hipocampal/enzimología , Región CA1 Hipocampal/crecimiento & desarrollo , Espinas Dendríticas/enzimología , Neuronas/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Sinapsis/enzimología , Animales , Animales Recién Nacidos , Región CA1 Hipocampal/citología , Células Cultivadas , Espinas Dendríticas/fisiología , Masculino , Neuronas/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Ratas , Ratas Sprague-Dawley , Sinapsis/fisiología
5.
Neurotherapeutics ; 19(5): 1566-1587, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35917088

RESUMEN

Sigma-1 receptor agonists have recently gained a great deal of interest due to their anti-amnesic, neuroprotective, and neurorestorative properties. Compounds such as PRE-084 or pridopidine (ACR16) are being studied as a potential treatment against cognitive decline associated with neurodegenerative disease, also to include Alzheimer's disease. Here, we performed in vitro experiments using primary neuronal cell cultures from rats to evaluate the abilities of ACR16 and PRE-084 to induce new synapses and spines formation, analyzing the expression of the possible genes and proteins involved. We additionally examined their neuroprotective properties against neuronal death mediated by oxidative stress and excitotoxicity. Both ACR16 and PRE-084 exhibited a concentration-dependent neuroprotective effect against NMDA- and H2O2-related toxicity, in addition to promoting the formation of new synapses and dendritic spines. However, only ACR16 generated dendritic spines involved in new synapse establishment, maintaining a more expanded activation of MAPK/ERK and PI3K/Akt signaling cascades. Consequently, ACR16 was also evaluated in vivo, and a dose of 1.5 mg/kg/day was administered intraperitoneally in APP/PS1 mice before performing the Morris water maze. ACR16 diminished the spatial learning and memory deficits observed in APP/PS1 transgenic mice via PI3K/Akt pathway activation. These data point to ACR16 as a pharmacological tool to prevent synapse loss and memory deficits associated with Alzheimer's disease, due to its neuroprotective properties against oxidative stress and excitotoxicity, as well as the promotion of new synapses and spines through a mechanism that involves AKT and ERK signaling pathways.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Fármacos Neuroprotectores , Ratones , Animales , Ratas , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fosfatidilinositol 3-Quinasas/farmacología , Fosfatidilinositol 3-Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/uso terapéutico , N-Metilaspartato/farmacología , N-Metilaspartato/uso terapéutico , Trastornos de la Memoria/metabolismo , Ratones Transgénicos , Modelos Animales de Enfermedad , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Aprendizaje por Laberinto
6.
PLoS One ; 10(3): e0118475, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25764078

RESUMEN

The PI3K-dependent activation of AKT results in the inhibition of GSK3ß in most signaling pathways. These kinases regulate multiple neuronal processes including the control of synapse number as shown for Drosophila and rodents. Alzheimer disease's patients exhibit high levels of circulating GSK3ß and, consequently, pharmacological strategies based on GSK3ß antagonists have been designed. The approach, however, has yielded inconclusive results so far. Here, we carried out a comparative study in Drosophila and rats addressing the role of GSK3ß in synaptogenesis. In flies, the genetic inhibition of the shaggy-encoded GSK3ß increases the number of synapses, while its upregulation leads to synapse loss. Likewise, in three weeks cultured rat hippocampal neurons, the pharmacological inhibition of GSK3ß increases synapse density and Synapsin expression. However, experiments on younger cultures (12 days) yielded an opposite effect, a reduction of synapse density. This unexpected finding seems to unveil an age- and dosage-dependent differential response of mammalian neurons to the stimulation/inhibition of GSK3ß, a feature that must be considered in the context of human adult neurogenesis and pharmacological treatments for Alzheimer's disease based on GSK3ß antagonists.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/citología , Neuronas/enzimología , Sinapsis/enzimología , Animales , Células Cultivadas , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/enzimología , Humanos , Neuronas/fisiología , Ratas , Transducción de Señal , Sinapsis/fisiología , Sinapsinas/metabolismo , Regulación hacia Arriba
7.
Front Mol Neurosci ; 6: 54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24427113

RESUMEN

PI3K activation promotes the formation of synaptic contacts and dendritic spines, morphological features of glutamatergic synapses that are commonly known to be related to learning processes. In this report, we show that in vivo administration of a peptide that activates the PI3K signaling pathway increases spine density in the rat hippocampus and enhances the animals' cognitive abilities, while in vivo electrophysiological recordings show that PI3K activation results in synaptic enhancement of Schaffer and stratum lacunosum moleculare inputs. Morphological characterization of the spines reveals that subjecting the animals to contextual fear-conditioning training per se promotes the formation of large spines, while PI3K activation reverts this effect and favors a general change toward small head areas. Studies using hippocampal neuronal cultures show that the PI3K spinogenic process is NMDA-dependent and activity-independent. In culture, PI3K activation was followed by mRNA upregulation of glutamate receptor subunits and of the immediate-early gene Arc. Time-lapse studies confirmed the ability of PI3K to induce the formation of small spines. Finally, we demonstrate that the spinogenic effect of PI3K can be induced in the presence of neurodegeneration, such as in the Tg2576 Alzheimer's mouse model. These findings highlight that the PI3K pathway is an important regulator of neuronal connectivity and stress the relationship between spine size and learning processes.

8.
Invest Ophthalmol Vis Sci ; 50(8): 3833-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19357356

RESUMEN

PURPOSE: It is well known that the small GTPase RhoA modulates actin cytoskeleton and cellular contractility in the trabecular meshwork (TM). Several substances known to contract the TM reduce outflow facility, whereas cellular relaxation is commonly associated with the opposite effect. Inhibitors of the RhoA pathway are under development as antiglaucoma drugs. Here the authors investigate the role of platelet-derived growth factor (PDGF), a known activator of the Rac1 pathway, in cell cytoskeleton, outflow facility, and intraocular pressure (IOP). METHODS: Effects of PDGF on actin cytoskeleton, Rac1, and AKT activation were tested in preconfluent and confluent bovine TM cells in culture. Rac1 and AKT/P-AKT activation were assessed by Western blot analysis. Trabecular outflow facility was measured in bovine perfused anterior segments. Changes in IOP were measured for up to 6 hours after topical application in the cornea of rabbit eyes by means of a contact tonometer. RESULTS: In TM cells, PDGF (10 ng/mL) activated Rac1 through AKT and induced actin cytoskeleton rearrangement with lamellipodia formation. In this sense, lamellipodia formation in TM cells was prevented by NSC23766, a Rac1 inhibitor, and LY294002, a PI3K inhibitor. In perfused anterior segments, PDGF (100 ng/mL) increased trabecular outflow facility by 26%. In vivo, when topically applied to rabbit corneas, PDGF induced a 20% decrease in IOP (100 ng/mL). This reduction was concentration dependent and presented an EC(50) value of 2.7 nM. CONCLUSIONS: PDGF, by activating the Rac1 pathway, induces cytoskeletal changes in TM cells that enhance outflow facility. Decreased IOP after PDGF application is likely caused by the facilitation of aqueous humor outflow. Rac1 pathway activation appears to be a positive modulator of outflow facility and an interesting target for decreasing IOP after ocular hypertension.


Asunto(s)
Humor Acuoso/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Malla Trabecular/efectos de los fármacos , Actinas/metabolismo , Administración Tópica , Animales , Segmento Anterior del Ojo/efectos de los fármacos , Segmento Anterior del Ojo/metabolismo , Western Blotting , Calcio/metabolismo , Bovinos , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Presión Intraocular/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Conejos , Tonometría Ocular , Malla Trabecular/citología , Malla Trabecular/metabolismo , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA