Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell Death Discov ; 10(1): 49, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38272891

RESUMEN

Electrical pulse stimulation has been used to enhance the differentiation or proliferation of neuronal progenitor cells in tissue engineering and cancer treatment. Therefore, a comprehensive investigation of the effects caused by its parameters is crucial for improvements in those fields. We propose a study of pulse parameters, to allow the control of N2a cell line fate and behavior. We have focused on designing an experimental setup that allows for the knowledge and control over the environment and the stimulation signals applied. To map the effects of the stimulation on N2a cells, their morphology and the cellular and molecular reactions induced by the pulse stimulation have been analyzed. Immunofluorescence, rt-PCR and western blot analysis have been carried out for this purpose, as well as cell counting. Our results show that low-amplitude electrical pulse stimulation promotes proliferation of N2a cells, whilst amplitudes in the range 250 mV/mm-500 mV/mm induce differentiation. Amplitudes higher than 750 mV/mm produce cell damage at low frequencies. For high frequencies, large amplitudes are needed to cause cell death. An inverse relation has been found between cell density and pulse-induced neuronal differentiation. The best condition for neuronal differentiation was found to be 500 mV/mm at 100 Hz. These findings have been confirmed by up-regulation of the Neurod1 gene. Our preliminary study of the molecular effects of electrical pulse stimulation on N2a offers premonitory clues of the PI3K/Akt/GSK-3ß pathway implications on the neuronal differentiation process through ES. In general, we have successfully mapped the sensitivity of N2a cells to electrical pulse stimulation parameters.

2.
Cells ; 13(2)2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-38247815

RESUMEN

The COVID-19 pandemic has brought to the forefront the intricate relationship between SARS-CoV-2 and its impact on neurological complications, including potential links to neurodegenerative processes, characterized by a dysfunction of the protein quality control systems and ER stress. This review article explores the role of protein quality control systems, such as the Unfolded Protein Response (UPR), the Endoplasmic Reticulum-Associated Degradation (ERAD), the Ubiquitin-Proteasome System (UPS), autophagy and the molecular chaperones, in SARS-CoV-2 infection. Our hypothesis suggests that SARS-CoV-2 produces ER stress and exploits the protein quality control systems, leading to a disruption in proteostasis that cannot be solved by the host cell. This disruption culminates in cell death and may represent a link between SARS-CoV-2 and neurodegeneration.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/complicaciones , Degradación Asociada con el Retículo Endoplásmico , Pandemias , Ubiquitina
3.
Healthcare (Basel) ; 12(16)2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39201232

RESUMEN

BACKGROUND: Cancer is a leading cause of morbidity and mortality globally. Dyspnea, affecting up to 60% of cancer patients, exacerbates physical and psychological distress, reducing quality of life. This study aims to explore the relationship between dyspnea and factors such as age, sex, clinical diagnosis, and treatment lines in cancer patients, with the goal of improving understanding and management of this debilitating symptom to enhance patient care and quality of life. METHODS: This study employed an observational, cross-sectional, and descriptive approach to investigate patients with oncological disease at the University Hospital of Salamanca between March 2021 and April 2024. A convenience sample was selected, including patients over 18 years old with a pathological diagnosis of cancer, experiencing any degree of dyspnea, and who consented to participate by signing the informed consent. Exclusion criteria included lack of consent and clinical conditions that prevented an interview. The studied variables encompass sociodemographic (age, gender, diagnosis, tumor stage, number of treatment lines) and clinical aspects (daily activities, degree of dyspnea, functional capacity, physical performance), evaluated using the Barthel Index, the mMRC Dyspnea Scale, the ECOG Scale, and the Short Physical Performance Battery (SPPB). Data were collected through semistructured interviews and medical records, and analyzed using specialized software. This research has ethical approval CEiM Code 2023 12 1472, Reference 2024/01. RESULTS: The mean age was 66.82 years. Lung cancer was predominant (60.2%), with most patients in stage 3 (65.7%) and receiving three treatment lines (68.7%). Higher age, advanced disease stage, and more treatment lines correlated with lower Barthel and SPPB scores, and higher ECOG and mMRC scores, indicating worse functionality, physical performance, and greater dyspnea. No significant correlations were found between gender or pathological diagnosis and the studied variables. CONCLUSIONS: Advanced age, higher disease stage, and more treatment lines are associated with decreased functionality, poorer physical performance, and increased dyspnea in cancer patients. Gender and specific cancer diagnosis do not significantly affect these relationships. Addressing dyspnea is crucial to improving the quality of life and physical performance in this population. Future studies should explore additional factors like treatment types and nutritional status.

4.
J Biol Chem ; 286(21): 18414-25, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21460223

RESUMEN

Neurotrophins, activating the PI3K/Akt signaling pathway, control neuronal survival and plasticity. Alterations in NGF, BDNF, IGF-1, or insulin signaling are implicated in the pathogenesis of Alzheimer disease. We have previously characterized a bigenic PS1×APP transgenic mouse displaying early hippocampal Aß deposition (3 to 4 months) but late (17 to 18 months) neurodegeneration of pyramidal cells, paralleled to the accumulation of soluble Aß oligomers. We hypothesized that PI3K/Akt/GSK-3ß signaling pathway could be involved in this apparent age-dependent neuroprotective/neurodegenerative status. In fact, our data demonstrated that, as compared with age-matched nontransgenic controls, the Ser-9 phosphorylation of GSK-3ß was increased in the 6-month PS1×APP hippocampus, whereas in aged PS1×APP animals (18 months), GSK-3ß phosphorylation levels displayed a marked decrease. Using N2a and primary neuronal cell cultures, we demonstrated that soluble amyloid precursor protein-α (sAPPα), the predominant APP-derived fragment in young PS1×APP mice, acting through IGF-1 and/or insulin receptors, activated the PI3K/Akt pathway, phosphorylated the GSK-3ß activity, and in consequence, exerted a neuroprotective action. On the contrary, several oligomeric Aß forms, present in the soluble fractions of aged PS1×APP mice, inhibited the induced phosphorylation of Akt/GSK-3ß and decreased the neuronal survival. Furthermore, synthetic Aß oligomers blocked the effect mediated by different neurotrophins (NGF, BDNF, insulin, and IGF-1) and sAPPα, displaying high selectivity for NGF. In conclusion, the age-dependent appearance of APP-derived soluble factors modulated the PI3K/Akt/GSK-3ß signaling pathway through the major neurotrophin receptors. sAPPα stimulated and Aß oligomers blocked the prosurvival signaling. Our data might provide insights into the selective vulnerability of specific neuronal groups in Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Envejecimiento/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Supervivencia Celular/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Transgénicos , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fosfatidilinositol 3-Quinasas/genética , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina
5.
J Neurochem ; 123(4): 635-44, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22913583

RESUMEN

Regulation of proteasome abundance to meet cell needs under stress conditions is critical for maintaining cellular homeostasis. However, the effects of aging on this homeostatic response remain unknown. In this report, we analyzed in young and aged rat hippocampus, the dynamics of proteasome recovery induced by proteasome stress. Proteasome inhibition in young rats leads to an early and coordinate transcriptional and translational up-regulation of both the catalytic subunits of constitutive proteasome and the proteasome maturation protein. By contrast, aged rats up-regulated the inducible catalytic subunits and showed a lower and shorter expression of proteasome maturation protein. This resulted in a faster recovery of proteasome activity in young rats. Importantly, proteasome inhibition highly affected pyramidal cells, leading to the accumulation of ubiquitinated proteins in perinuclear regions of aged, but not young pyramidal neurons. These data strongly suggest that age-dependent differences in proteasome level and composition could contribute to neurodegeneration induced by proteasome dysfunction in normal and pathological aging.


Asunto(s)
Envejecimiento , Hipocampo/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Regulación hacia Arriba/fisiología , Factores de Edad , Animales , Dominio Catalítico/fisiología , Nucléolo Celular/metabolismo , Hipocampo/citología , Inmunoproteínas/metabolismo , Masculino , Neuronas/citología , Neuronas/metabolismo , Ratas , Ratas Wistar
6.
J Neuroinflammation ; 9: 87, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22559833

RESUMEN

BACKGROUND: Neuroinflammation and protein accumulation are characteristic hallmarks of both normal aging and age-related neurodegenerative diseases. However, the relationship between these factors in neurodegenerative processes is poorly understood. We have previously shown that proteasome inhibition produced higher neurodegeneration in aged than in young rats, suggesting that other additional age-related events could be involved in neurodegeneration. We evaluated the role of lipopolysaccharide (LPS)-induced neuroinflammation as a potential synergic risk factor for hippocampal neurodegeneration induced by proteasome inhibition. METHODS: Young male Wistar rats were injected with 1 µL of saline or LPS (5 mg/mL) into the hippocampus to evaluate the effect of LPS-induced neuroinflammation on protein homeostasis. The synergic effect of LPS and proteasome inhibition was analyzed in young rats that first received 1 µL of LPS and 24 h later 1 µL (5 mg/mL) of the proteasome inhibitor lactacystin. Animals were sacrificed at different times post-injection and hippocampi isolated and processed for gene expression analysis by real-time polymerase chain reaction; protein expression analysis by western blots; proteasome activity by fluorescence spectroscopy; immunofluorescence analysis by confocal microscopy; and degeneration assay by Fluoro-Jade B staining. RESULTS: LPS injection produced the accumulation of ubiquitinated proteins in hippocampal neurons, increased expression of the E2 ubiquitin-conjugating enzyme UB2L6, decreased proteasome activity and increased immunoproteasome content. However, LPS injection was not sufficient to produce neurodegeneration. The combination of neuroinflammation and proteasome inhibition leads to higher neuronal accumulation of ubiquitinated proteins, predominant expression of pro-apoptotic markers and increased neurodegeneration, when compared with LPS or lactacystin (LT) injection alone. CONCLUSIONS: Our results identify neuroinflammation as a risk factor that increases susceptibility to neurodegeneration induced by proteasome inhibition. These results highlight the modulation of neuroinflammation as a mechanism for neuronal protection that could be relevant in situations where both factors are present, such as aging and neurodegenerative diseases.


Asunto(s)
Hipocampo/efectos de los fármacos , Lipopolisacáridos/toxicidad , Degeneración Nerviosa/inducido químicamente , Inhibidores de Proteasoma/toxicidad , Acetilcisteína/análogos & derivados , Acetilcisteína/toxicidad , Envejecimiento/efectos de los fármacos , Envejecimiento/fisiología , Animales , Sinergismo Farmacológico , Hipocampo/enzimología , Hipocampo/patología , Inflamación/inducido químicamente , Inflamación/epidemiología , Inflamación/patología , Masculino , Degeneración Nerviosa/epidemiología , Degeneración Nerviosa/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Ratas Wistar , Ubiquitinación/efectos de los fármacos , Ubiquitinación/fisiología
7.
Acta Neuropathol ; 123(1): 53-70, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22020633

RESUMEN

Dystrophic neurites associated with amyloid plaques precede neuronal death and manifest early in Alzheimer's disease (AD). In this work we have characterized the plaque-associated neuritic pathology in the hippocampus of young (4- to 6-month-old) PS1(M146L)/APP(751SL) mice model, as the initial degenerative process underlying functional disturbance prior to neuronal loss. Neuritic plaques accounted for almost all fibrillar deposits and an axonal origin of the dystrophies was demonstrated. The early induction of autophagy pathology was evidenced by increased protein levels of the autophagosome marker LC3 that was localized in the axonal dystrophies, and by electron microscopic identification of numerous autophagic vesicles filling and causing the axonal swellings. Early neuritic cytoskeletal defects determined by the presence of phosphorylated tau (AT8-positive) and actin-cofilin rods along with decreased levels of kinesin-1 and dynein motor proteins could be responsible for this extensive vesicle accumulation within dystrophic neurites. Although microsomal Aß oligomers were identified, the presence of A11-immunopositive Aß plaques also suggested a direct role of plaque-associated Aß oligomers in defective axonal transport and disease progression. Most importantly, presynaptic terminals morphologically disrupted by abnormal autophagic vesicle buildup were identified ultrastructurally and further supported by synaptosome isolation. Finally, these early abnormalities in axonal and presynaptic structures might represent the morphological substrate of hippocampal dysfunction preceding synaptic and neuronal loss and could significantly contribute to AD pathology in the preclinical stages.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Axones/ultraestructura , Vesículas Citoplasmáticas/ultraestructura , Hipocampo/ultraestructura , Neuritas/ultraestructura , Placa Amiloide/ultraestructura , Enfermedad de Alzheimer/metabolismo , Animales , Autofagia , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Microscopía Inmunoelectrónica , Neuronas/metabolismo , Placa Amiloide/metabolismo , Terminales Presinápticos/ultraestructura
8.
Front Mol Biosci ; 8: 658742, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34222330

RESUMEN

Aging is a biological and multifactorial process characterized by a progressive and irreversible deterioration of the physiological functions leading to a progressive increase in morbidity. In the next decades, the world population is expected to reach ten billion, and globally, elderly people over 80 are projected to triple in 2050. Consequently, it is also expected an increase in the incidence of age-related pathologies such as cancer, diabetes, or neurodegenerative disorders. Disturbance of cellular protein homeostasis (proteostasis) is a hallmark of normal aging that increases cell vulnerability and might be involved in the etiology of several age-related diseases. This review will focus on the molecular alterations occurring during normal aging in the most relevant protein quality control systems such as molecular chaperones, the UPS, and the ALS. Also, alterations in their functional cooperation will be analyzed. Finally, the role of inflammation, as a synergistic negative factor of the protein quality control systems during normal aging, will also be addressed. A better comprehension of the age-dependent modifications affecting the cellular proteostasis, as well as the knowledge of the mechanisms underlying these alterations, might be very helpful to identify relevant risk factors that could be responsible for or contribute to cell deterioration, a fundamental question still pending in biomedicine.

9.
Aging Cell ; 19(11): e13257, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33146912

RESUMEN

Diabetes and metabolic syndrome are associated with the typical American high glycemia diet and result in accumulation of high levels of advanced glycation end products (AGEs), particularly upon aging. AGEs form when sugars or their metabolites react with proteins. Associated with a myriad of age-related diseases, AGEs accumulate in many tissues and are cytotoxic. To date, efforts to limit glycation pharmacologically have failed in human trials. Thus, it is crucial to identify systems that remove AGEs, but such research is scanty. Here, we determined if and how AGEs might be cleared by autophagy. Our in vivo mouse and C. elegans models, in which we altered proteolysis or glycative burden, as well as experiments in five types of cells, revealed more than six criteria indicating that p62-dependent autophagy is a conserved pathway that plays a critical role in the removal of AGEs. Activation of autophagic removal of AGEs requires p62, and blocking this pathway results in accumulation of AGEs and compromised viability. Deficiency of p62 accelerates accumulation of AGEs in soluble and insoluble fractions. p62 itself is subject to glycative inactivation and accumulates as high mass species. Accumulation of p62 in retinal pigment epithelium is reversed by switching to a lower glycemia diet. Since diminution of glycative damage is associated with reduced risk for age-related diseases, including age-related macular degeneration, cardiovascular disease, diabetes, Alzheimer's, and Parkinson's, discovery of methods to limit AGEs or enhance p62-dependent autophagy offers novel potential therapeutic targets to treat AGEs-related pathologies.


Asunto(s)
Productos Finales de Glicación Avanzada/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Autofagia/fisiología , Línea Celular , Supervivencia Celular/fisiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Riñón/citología , Riñón/metabolismo , Cristalino/citología , Cristalino/metabolismo , Lisosomas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas
10.
J Neurosci ; 28(45): 11650-61, 2008 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-18987201

RESUMEN

Although the microglial activation is concomitant to the Alzheimer's disease, its precise role (neuroprotection vs neurodegeneration) has not yet been resolved. Here, we show the existence of an age-dependent phenotypic change of microglial activation in the hippocampus of PS1xAPP model, from an alternative activation state with Abeta phagocytic capabilities (at 6 months) to a classic cytotoxic phenotype (expressing TNF-alpha and related factors) at 18 months of age. This switch was coincident with high levels of soluble Abeta oligomers and a significant pyramidal neurodegeneration. In vitro assays, using astromicroglial cultures, demonstrated that oligomeric Abeta42 and soluble extracts from 18-month-old PS1xAPP hippocampus produced a potent TNF-alpha induction whereas monomeric Abeta42 and soluble extract from 6- or 18-month-old control and 6-month-old PS1xAPP hippocampi produced no stimulation. This stimulatory effect was avoided by immunodepletion using 6E10 or A11. In conclusion, our results show evidence of a switch in the activated microglia phenotype from alternative, at the beginning of Abeta pathology, to a classical at advanced stage of the disease in this model. This change was induced, at least in part, by the age-dependent accumulation of extracellular soluble Abeta oligomers. Finally, these cytotoxic activated microglial cells could participate in the neuronal lost observed in AD.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Inflamación/etiología , Microglía/patología , Mutación/genética , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Antígeno CD11b/metabolismo , Complejo CD3/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Lectinas/metabolismo , Ratones , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Presenilina-1/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
J Neurochem ; 108(1): 260-72, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19012754

RESUMEN

Alterations in the proteasome activity in the CNS have been described during aging. However, a detailed study of all proteasome subunits is actually lacking. We have analyzed, in vivo, the age-related modifications in the molecular composition of hippocampal proteasomes. We found that the immunoproteasome/proteasome ratio was increased in aged hippocampus. The processing of the low-molecular-mass protein (LMP)7/beta(5i) subunit, practically absent in young hippocampus, was increased in aged animals. Among the potential factors underlying these modifications we evaluated the neuroinflammation and the transcription factor Zif268. Lipopolysaccharide (LPS)-induced neuroinflammation in young rats, up-regulated the expression of immunoproteasome subunits and increased the processing of the LMP7/beta(5i) protein. Moreover, the hydrophobicity of cellular peptides, analyzed by liquid chromatography, increased in both, young LPS-injected animals and aged rats, suggesting that immunoproteasomes including the LMP7/beta(5i) subunit could, at least in part, account for this modification. Also, the mRNA expression of the transcription factor Zif268, which down-regulates the immunoproteasome subunit LMP7/beta(5i) by binding to sequences within the promoter regions, was decreased in both, aged hippocampus and young LPS-injected animals. Finally, we found that spatial memory training in young animals, a situation in which the expression of Zif268 is increased, modified the mRNA expression of the constitutive and catalytic subunits in an opposite manner. Based on present data, we propose that the age-related increases in the content of hippocampal immunoproteasome is mostly because of neuroinflammatory processes associated to aging.


Asunto(s)
Envejecimiento/metabolismo , Regulación de la Expresión Génica/fisiología , Hipocampo/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/ultraestructura , Inmunoglobulinas/metabolismo , Lipopolisacáridos/farmacología , Masculino , Aprendizaje por Laberinto/fisiología , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Fracciones Subcelulares/metabolismo
12.
J Chem Neuroanat ; 35(1): 12-32, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17616449

RESUMEN

The nitrergic system has been inferred from cells positive to nicotinamide adenine dinucleotide phosphate diaphorase (NADPHd) histochemistry and/or to the neuronal isoform of nitric oxide synthase (nNOS) immunohistochemistry in different species of vertebrates. The aim of the present work was to systematically study the distribution of cell producing nitric oxide in the goldfish (Carassius auratus) brain. To reach this goal, we firstly studied co-localization for NADPHd and nNOS techniques and demonstrated an extensive double labeling. Then, we studied the distribution through the brain by the two separate methods and found labeled cells widely distributed in brain and spinal cord. In the telencephalon, such cells were in both dorsal and ventral areas. In the diencephalon, the cells were found in some nuclei of the preoptic area and hypothalamus, habenula, pretectum, and dorsal and ventral thalamic regions. In the midbrain, cells were observed in the optic tectum, torus longitudinalis, and tegmental nuclei. In the rhombencephalon, cells were found in the cerebellum, the reticular formation, the locus coeruleus, the raphe nuclei, and the nuclei of the cranial nerves. Labeled cells were also observed in the gray area of the spinal cord. Cognizing that a direct comparison of the present results with those reported in other vertebrates is not clear-cut because of homologies; we conclude that the nitrergic system is roughly similar from fish to mammals.


Asunto(s)
Sistema Nervioso Central/enzimología , Carpa Dorada/metabolismo , NADPH Deshidrogenasa/metabolismo , Neuronas Nitrérgicas/enzimología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/biosíntesis , Animales , Evolución Biológica , Encéfalo/anatomía & histología , Encéfalo/enzimología , Mapeo Encefálico , Sistema Nervioso Central/anatomía & histología , Femenino , Carpa Dorada/anatomía & histología , Histocitoquímica , Inmunohistoquímica , Masculino , Neuronas Nitrérgicas/citología , Especificidad de la Especie , Médula Espinal/anatomía & histología , Médula Espinal/enzimología
13.
Cell Death Dis ; 9(5): 531, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29748539

RESUMEN

Sirtuin 1 (SIRT1) activity is neuroprotective, and we have recently demonstrated its role in the retrograde degenerative process in motoneurons (MNs) in the spinal cord of rats after peripheral nerve root avulsion (RA) injury. SIRT2 has been suggested to exert effects opposite those of SIRT1; however, its roles in neurodegeneration and neuron response after nerve injury remain unclear. Here we compared the neuroprotective potentials of SIRT1 activation and SIRT2 inhibition in a mouse model of hypoglossal nerve axotomy. This injury induced a reduction of around half MN population within the hypoglossal nucleus by a non-apoptotic neurodegenerative process triggered by endoplasmic reticulum (ER) stress that resulted in activation of the unfolded protein response mediated by IRE1α and XBP1 by 21 days post injury. Both SIRT1 activation with NeuroHeal and SIRT2 inhibition with AK7 protected NSC-34 motor neuron-like cells against ER stress in vitro. In agreement with the in vitro results, NeuroHeal treatment or SIRT1 overexpression was neuroprotective of axotomized hypoglossal MNs in a transgenic mouse model. In contrast, AK7 treatment or SIRT2 genetic depletion in mice inhibited damaged MN survival. To resolve the in vitro/in vivo discrepancies, we used an organotypic spinal cord culture system that preserves glial cells. In this system, AK7 treatment of ER-stressed organotypic cultures was detrimental for MNs and increased microglial nuclear factor-κB and the consequent transcription of cytotoxic pro-inflammatory factors similarly. The results highlight the importance of glial cells in determining the neuroprotective impact of any treatment.


Asunto(s)
Acamprosato/farmacología , Benzamidas/farmacología , Traumatismos del Nervio Hipogloso , Neuronas Motoras/enzimología , Neuroprotección/efectos de los fármacos , Ribavirina/farmacología , Sirtuina 1 , Sirtuina 2 , Sulfonamidas/farmacología , Animales , Combinación de Medicamentos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Femenino , Traumatismos del Nervio Hipogloso/tratamiento farmacológico , Traumatismos del Nervio Hipogloso/enzimología , Traumatismos del Nervio Hipogloso/genética , Traumatismos del Nervio Hipogloso/patología , Ratones , Ratones Noqueados , Neuronas Motoras/patología , Neuroprotección/genética , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Sirtuina 1/metabolismo , Sirtuina 2/antagonistas & inhibidores , Sirtuina 2/genética , Sirtuina 2/metabolismo
14.
Sci Rep ; 7(1): 8100, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28808322

RESUMEN

Proteostasis alteration and neuroinflammation are typical features of normal aging. We have previously shown that neuroinflammation alters cellular proteostasis through immunoproteasome induction, leading to a transient decrease of proteasome activity. Here, we further investigated the role of acute lipopolysaccharide (LPS)-induced hippocampal neuroinflammation in cellular proteostasis. In particular, we focused on macroautophagy (hereinafter called autophagy) and endoplasmic reticulum-associated protein degradation (ERAD). We demonstrate that LPS injection induced autophagy activation that was dependent, at least in part, on glycogen synthase kinase (GSK)-3ß activity but independent of mammalian target of rapamycin (mTOR) inhibition. Neuroinflammation also produced endoplasmic reticulum (ER) stress leading to canonical unfolded protein response (UPR) activation with a rapid activating transcription factor (ATF) 6α attenuation that resulted in a time-dependent down-regulation of ERAD markers. In this regard, the time-dependent accumulation of unspliced X-box binding protein (XBP) 1, likely because of decreased inositol-requiring enzyme (IRE) 1α-mediated splicing activity, might underlie in vivo ATF6α attenuation. Importantly, lactacystin-induced activation of ERAD was abolished in both the acute neuroinflammation model and in aged rats. Therefore, we provide a cellular pathway through which neuroinflammation might sensitize cells to neurodegeneration under stress situations, being relevant in normal aging and other disorders where neuroinflammation is a characteristic feature.


Asunto(s)
Autofagia/fisiología , Estrés del Retículo Endoplásmico/fisiología , Degradación Asociada con el Retículo Endoplásmico/fisiología , Inflamación/fisiopatología , Proteostasis/fisiología , Factor de Transcripción Activador 6/metabolismo , Animales , Línea Celular , Regulación hacia Abajo/fisiología , Endorribonucleasas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Respuesta de Proteína Desplegada/fisiología , Proteína 1 de Unión a la X-Box/metabolismo
15.
Neurobiol Aging ; 27(7): 973-82, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15964666

RESUMEN

Aging represents the main risk factor to develop Alzheimer disease (AD) and protein aggregation constitutes a pathological hallmark thought to be involved in the etiology of this disease. Here, we show that, in basal conditions, the expression of chaperones calnexin, protein disulfide isomerase (PDI) and Grp78 was decreased in aged hippocampus, whereas the protein ubiquitination increased, suggesting the existence of age-related deficits in the systems involved in the defense against unfolded proteins. Interestingly, when cellular stress was induced by intra-hippocampal lactacystin injection, the aged rats were less efficient than young animals in alleviating the protein accumulation and, as an important factor, did not induce the expression of chaperones as young animals. However, the expression of the pro-apoptotic factor CHOP/GADD153 was induced and caspase-12 was activated in stressed aged rats but not in young animals. Current results demonstrated that unfolding protein response (UPR) is not correctly activated in aged rat hippocampus. Consequently, the up-regulation of apoptotic pathway mediators is increased in aged rats. Results might provide further understanding of the pathogenic mechanisms of age-related neurodegenerative disorders.


Asunto(s)
Envejecimiento/metabolismo , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Estrés Fisiológico/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Apoptosis/fisiología , Calnexina/metabolismo , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/metabolismo , Hipocampo/patología , Masculino , Chaperonas Moleculares/metabolismo , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Pliegue de Proteína , Ratas , Transducción de Señal/fisiología , Estrés Fisiológico/fisiopatología , Factor de Transcripción CHOP/metabolismo , Ubiquitina/metabolismo , Regulación hacia Arriba/fisiología
16.
Sci Rep ; 5: 10027, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25941117

RESUMEN

Targeting the ubiquitin proteasome pathway has emerged as a rational approach in the treatment of human cancers. Autophagy has been described as a cytoprotective mechanism to increase tumor cell survival under stress conditions. Here, we have focused on the role of proteasome inhibition in cell cycle progression and the role of autophagy in the proliferation recovery. The study was performed in the breast cancer cell line MCF7 compared to the normal mammary cell line MCF10A. We found that the proteasome inhibitor MG132 induced G1/S arrest in MCF10A, but G2/M arrest in MCF7 cells. The effect of MG132 on MCF7 was reproduced on MCF10A cells in the presence of the glycogen synthase kinase 3ß (GSK-3ß) inhibitor VII. Similarly, MCF7 cells overexpressing constitutively active GSK-3ß behaved like MCF10A cells. On the other hand, MCF10A cells remained arrested after MG132 removal while MCF7 recovered the proliferative capacity. Importantly, this recovery was abolished in the presence of the autophagy inhibitor 3-methyladenine (3-MA). Thus, our results support the relevance of GSK-3ß and autophagy as two targets for controlling cell cycle progression and proliferative capacity in MCF7, highlighting the co-treatment of breast cancer cells with 3-MA to synergize the effect of the proteasome inhibition.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Puntos de Control de la Fase G1 del Ciclo Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Puntos de Control de la Fase S del Ciclo Celular , Autofagia , Proliferación Celular , Glucógeno Sintasa Quinasa 3 beta , Humanos , Células MCF-7 , Transducción de Señal
17.
Neurobiol Aging ; 36(5): 1953-63, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25817083

RESUMEN

Autophagy plays a key role in the maintenance of cellular homeostasis, and autophagy deregulation gives rise to severe disorders. Many of the signaling pathways regulating autophagy under stress conditions are still poorly understood. Using a model of proteasome stress in rat hippocampus, we have characterized the functional crosstalk between the ubiquitin proteasome system and the autophagy-lysosome pathway, identifying also age-related modifications in the crosstalk between both proteolytic systems. Under proteasome inhibition, both autophagy activation and resolution were efficiently induced in young but not in aged rats, leading to restoration of protein homeostasis only in young pyramidal neurons. Importantly, proteasome stress inhibited glycogen synthase kinase-3ß in young but activated in aged rats. This age-related difference could be because of a dysfunction in the signaling pathway of the insulin growth factor-1 under stress situations. Present data highlight the potential role of glycogen synthase kinase-3ß in the coordination of both proteolytic systems under stress situation, representing a key molecular target to sort out this deleterious effect.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/fisiología , Autofagia/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Hipocampo/fisiología , Lisosomas/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Células Piramidales/metabolismo , Transducción de Señal/fisiología , Animales , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Homeostasis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Enfermedades Neurodegenerativas/genética , Inhibidores de Proteasoma , Proteínas/metabolismo , Proteolisis , Células Piramidales/fisiología , Ratas Wistar , Ubiquitina/fisiología
18.
Brain Res Dev Brain Res ; 148(1): 129-41, 2004 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-14757527

RESUMEN

Here we have studied the developmental expression of alpha1 subunit of the GABAA receptor in comparison with the expression of alpha2 subunit and several GABAergic markers (parvalbumin (PV), calretinin (CR), somatostatin (SOM), neuropeptide Y (NPY) and vasoactive intestinal polypeptide (VIP)). The alpha1 expression (mRNA and protein) was low at birth and increased progressively until the adulthood. This expression pattern was similar to that observed for PV, opposite to that of CR (high at birth and decreased continuously until the adulthood) and differed from that observed for the alpha2 and neuropeptides (SOM, NPY and VIP) (in all cases, a clear peak in expression was observed at P10). We further investigated the expression of alpha1, PV and CR by immunohistochemistry. As expected, the alpha1 and the PV expression were low at birth and increased progressively until the adulthood. Both alpha1 and PV were co-expressed by the same interneuronal population, however, the maturation of the alpha1 subunit preceded to that of PV. Finally, we observed a gradient of maturation between the different fields of the hippocampus proper (CA2-3 preceded to CA1 and DG). This gradient could be related to the high expression of CR positive cells and fibers during the first 10 postnatal days, located principally in the stratum lacunosum moleculare of the CA2-3 layers.


Asunto(s)
Envejecimiento/metabolismo , Expresión Génica , Hipocampo/metabolismo , Subunidades de Proteína/metabolismo , Receptores de GABA-A/metabolismo , Animales , Animales Recién Nacidos , Western Blotting/métodos , Calbindina 2 , Hipocampo/crecimiento & desarrollo , Inmunohistoquímica/métodos , Neuropéptido Y/metabolismo , Parvalbúminas/metabolismo , Subunidades de Proteína/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Receptores de GABA-A/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Proteína G de Unión al Calcio S100/metabolismo , Somatostatina/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
19.
Brain Res Dev Brain Res ; 151(1-2): 87-98, 2004 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-15246695

RESUMEN

The GABAergic system plays an important role in the hippocampal development. Here we have studied the developmental expression of the alpha 5 subunit of the GABA(A) receptor (from rat hippocampus) by RT-competitive PCR, immunoblot and immunocytochemistry. Our results demonstrated an early induction of the alpha 5 subunit expression (at mRNA and protein levels) during the first postnatal week, peaking at P5 and decreasing after this age. The peak of alpha 5 subunit expression precedes the peak of expression for the synaptophysin, GAD65 and GAD67. Thus, the increase in the alpha 5 GABA(A) receptor subunit expression may precede the GABAergic synaptogenesis. Importantly, between P0 and P7, the expression of the alpha 5 subunit was concentrated at the cell somata of the pyramidal and granular cells. After P10, its localization shifted from the cell bodies to the dendritic layers. This developmental pattern is similar to that reported for the Na(+)-K(+)-2Cl(-) system and it might be correlated with the transition from excitatory to inhibitory GABAergic activity.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/metabolismo , Subunidades de Proteína/metabolismo , Receptores de GABA-A/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Western Blotting/métodos , Recuento de Células/métodos , Glutamato Descarboxilasa/metabolismo , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Inmunohistoquímica/métodos , Isoenzimas/metabolismo , Neuronas/metabolismo , Subunidades de Proteína/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Receptores de GABA-A/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sinaptofisina/metabolismo
20.
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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA