Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biol Res ; 57(1): 39, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38867288

RESUMEN

BACKGROUND: Spreading depression (SD) is an intriguing phenomenon characterized by massive slow brain depolarizations that affect neurons and glial cells. This phenomenon is repetitive and produces a metabolic overload that increases secondary damage. However, the mechanisms associated with the initiation and propagation of SD are unknown. Multiple lines of evidence indicate that persistent and uncontrolled opening of hemichannels could participate in the pathogenesis and progression of several neurological disorders including acute brain injuries. Here, we explored the contribution of astroglial hemichannels composed of connexin-43 (Cx43) or pannexin-1 (Panx1) to SD evoked by high-K+ stimulation in brain slices. RESULTS: Focal high-K+ stimulation rapidly evoked a wave of SD linked to increased activity of the Cx43 and Panx1 hemichannels in the brain cortex, as measured by light transmittance and dye uptake analysis, respectively. The activation of these channels occurs mainly in astrocytes but also in neurons. More importantly, the inhibition of both the Cx43 and Panx1 hemichannels completely prevented high K+-induced SD in the brain cortex. Electrophysiological recordings also revealed that Cx43 and Panx1 hemichannels critically contribute to the SD-induced decrease in synaptic transmission in the brain cortex and hippocampus. CONCLUSIONS: Targeting Cx43 and Panx1 hemichannels could serve as a new therapeutic strategy to prevent the initiation and propagation of SD in several acute brain injuries.


Asunto(s)
Astrocitos , Conexina 43 , Conexinas , Depresión de Propagación Cortical , Transmisión Sináptica , Animales , Astrocitos/fisiología , Conexinas/metabolismo , Depresión de Propagación Cortical/fisiología , Depresión de Propagación Cortical/efectos de los fármacos , Transmisión Sináptica/fisiología , Transmisión Sináptica/efectos de los fármacos , Conexina 43/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Corteza Cerebral , Neuronas/fisiología , Hipocampo , Ratas Sprague-Dawley , Ratas , Potasio/metabolismo
2.
J Neuroinflammation ; 21(1): 144, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822334

RESUMEN

Cumulative evidence has established that Interferon (IFN)-γ has both pathogenic and protective roles in Multiple Sclerosis and the animal model, Experimental Autoimmune Encephalomyelitis (EAE). However, the underlying mechanisms to the beneficial effects of IFN-γ are not well understood. In this study, we found that IFN-γ exerts therapeutic effects on chronic, relapsing-remitting, and chronic progressive EAE models. The frequency of regulatory T (Treg) cells in spinal cords from chronic EAE mice treated with IFN-γ was significantly increased with no effect on Th1 and Th17 cells. Consistently, depletion of FOXP3-expressing cells blocked the protective effects of IFN-γ, indicating that the therapeutic effect of IFN-γ depends on the presence of Treg cells. However, IFN-γ did not trigger direct in vitro differentiation of Treg cells. In vivo administration of blocking antibodies against either interleukin (IL)-10, transforming growth factor (TGF)-ß or program death (PD)-1, revealed that the protective effects of IFN-γ in EAE were also dependent on TGF-ß and PD-1, but not on IL-10, suggesting that IFN-γ might have an indirect role on Treg cells acting through antigen-presenting cells. Indeed, IFN-γ treatment increased the frequency of a subset of splenic CD11b+ myeloid cells expressing TGF-ß-Latency Associated Peptide (LAP) and program death ligand 1 (PD-L1) in a signal transducer and activator of transcription (STAT)-1-dependent manner. Furthermore, splenic CD11b+ cells from EAE mice preconditioned in vitro with IFN-γ and myelin oligodendrocyte glycoprotein (MOG) peptide exhibited a tolerogenic phenotype with the capability to induce conversion of naïve CD4+ T cells mediated by secretion of TGF-ß. Remarkably, adoptive transfer of splenic CD11b+ cells from IFN-γ-treated EAE mice into untreated recipient mice ameliorated clinical symptoms of EAE and limited central nervous system infiltration of mononuclear cells and effector helper T cells. These results reveal a novel cellular and molecular mechanism whereby IFN-γ promotes beneficial effects in EAE by endowing splenic CD11b+ myeloid cells with tolerogenic and therapeutic activities.


Asunto(s)
Antígeno CD11b , Encefalomielitis Autoinmune Experimental , Interferón gamma , Ratones Endogámicos C57BL , Células Mieloides , Bazo , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Ratones , Interferón gamma/metabolismo , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Bazo/inmunología , Antígeno CD11b/metabolismo , Femenino , Glicoproteína Mielina-Oligodendrócito/toxicidad , Glicoproteína Mielina-Oligodendrócito/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Factores de Transcripción Forkhead/metabolismo , Modelos Animales de Enfermedad
3.
Front Immunol ; 14: 1191838, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37334380

RESUMEN

Compelling evidence has shown that interferon (IFN)-γ has dual effects in multiple sclerosis and in its animal model of experimental autoimmune encephalomyelitis (EAE), with results supporting both a pathogenic and beneficial function. However, the mechanisms whereby IFN-γ may promote neuroprotection in EAE and its effects on central nervous system (CNS)-resident cells have remained an enigma for more than 30 years. In this study, the impact of IFN-γ at the peak of EAE, its effects on CNS infiltrating myeloid cells (MC) and microglia (MG), and the underlying cellular and molecular mechanisms were investigated. IFN-γ administration resulted in disease amelioration and attenuation of neuroinflammation associated with significantly lower frequencies of CNS CD11b+ myeloid cells and less infiltration of inflammatory cells and demyelination. A significant reduction in activated MG and enhanced resting MG was determined by flow cytometry and immunohistrochemistry. Primary MC/MG cultures obtained from the spinal cord of IFN-γ-treated EAE mice that were ex vivo re-stimulated with a low dose (1 ng/ml) of IFN-γ and neuroantigen, promoted a significantly higher induction of CD4+ regulatory T (Treg) cells associated with increased transforming growth factor (TGF)-ß secretion. Additionally, IFN-γ-treated primary MC/MG cultures produced significantly lower nitrite in response to LPS challenge than control MC/MG. IFN-γ-treated EAE mice had a significantly higher frequency of CX3CR1high MC/MG and expressed lower levels of program death ligand 1 (PD-L1) than PBS-treated mice. Most CX3CR1highPD-L1lowCD11b+Ly6G- cells expressed MG markers (Tmem119, Sall2, and P2ry12), indicating that they represented an enriched MG subset (CX3CR1highPD-L1low MG). Amelioration of clinical symptoms and induction of CX3CR1highPD-L1low MG by IFN-γ were dependent on STAT-1. RNA-seq analyses revealed that in vivo treatment with IFN-γ promoted the induction of homeostatic CX3CR1highPD-L1low MG, upregulating the expression of genes associated with tolerogenic and anti-inflammatory roles and down-regulating pro-inflammatory genes. These analyses highlight the master role that IFN-γ plays in regulating microglial activity and provide new insights into the cellular and molecular mechanisms involved in the therapeutic activity of IFN-γ in EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Ratones , Animales , Microglía/metabolismo , Interferón gamma/metabolismo , Antígeno B7-H1/metabolismo , Sistema Nervioso Central
4.
iScience ; 24(12): 103478, 2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34841222

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly rampaged worldwide, causing a pandemic of coronavirus disease (COVID -19), but the biology of SARS-CoV-2 remains under investigation. We demonstrate that both SARS-CoV-2 spike protein and human coronavirus 229E (hCoV-229E) or its purified S protein, one of the main viruses responsible for the common cold, induce the transient opening of Pannexin-1 (Panx-1) channels in human lung epithelial cells. However, the Panx-1 channel opening induced by SARS-CoV-2 is greater and more prolonged than hCoV-229E/S protein, resulting in an enhanced ATP, PGE2, and IL-1ß release. Analysis of lung lavages and tissues indicate that Panx-1 mRNA expression is associated with increased ATP, PGE2, and IL-1ß levels. Panx-1 channel opening induced by SARS-CoV-2 spike protein is angiotensin-converting enzyme 2 (ACE-2), endocytosis, and furin dependent. Overall, we demonstrated that Panx-1 channel is a critical contributor to SARS-CoV-2 infection and should be considered as an alternative therapy.

5.
Biomed Res Int ; 2021: 6614000, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34337036

RESUMEN

Chloroquine (CQ) and hydroxychloroquine (HCQ) have shown the ability to inhibit in vitro viral replications of coronaviridae viruses such as SARS-CoV and SARS-CoV-2. However, clinical trial outcomes have been disparate, suggesting that CQ and HCQ antiviral mechanisms are not fully understood. Based on three-dimensional structural similarities between HCQ and the known ACE2 specific inhibitor MLN-4760, we compared their modulation on ACE2 activity. Here we describe, for the first time, in a cell-free in vitro system that HCQ directly and dose-dependently inhibits the activity of recombinant human ACE2, with a potency similar to the MLN-4760. Further analysis suggests that HCQ binds to a noncompetitive site other than the one occupied by MLN-4760. We also determined that the viral spike glycoprotein segment that comprises the RBD segment has no effect on ACE2 activity but unexpectedly was able to partially reverse the inhibition induced by HCQ but not that by MLN-4760. In summary, here we demonstrate the direct inhibitory action of HCQ over the activity of the enzyme ACE2. Then, by determining the activity of ACE2, we reveal that the interaction with the spike protein of SARS-CoV-2 leads to structural changes that at least partially displace the interaction of the said enzyme with HCQ. These results may help to explain why the effectiveness of HCQ in clinical trials has been so variable. Additionally, this knowledge could be used for to develop techniques for the detection of SARS-CoV-2.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Antivirales , Tratamiento Farmacológico de COVID-19 , Hidroxicloroquina , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Humanos , Hidroxicloroquina/química , Hidroxicloroquina/metabolismo , Hidroxicloroquina/farmacología , Imidazoles/química , Imidazoles/metabolismo , Imidazoles/farmacología , Leucina/análogos & derivados , Leucina/química , Leucina/metabolismo , Leucina/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo
6.
PLoS One ; 12(6): e0177472, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28650992

RESUMEN

Galectin-8 (Gal-8) is a member of a glycan-binding protein family that regulates the immune system, among other functions, and is a target of antibodies in autoimmune disorders. However, its role in multiple sclerosis (MS), an autoimmune inflammatory disease of the central nervous system (CNS), remains unknown. We study the consequences of Gal-8 silencing on lymphocyte subpopulations and the development of experimental autoimmune encephalitis (EAE), to then assess the presence and clinical meaning of anti-Gal-8 antibodies in MS patients. Lgals8/Lac-Z knock-in mice lacking Gal-8 expression have higher polarization toward Th17 cells accompanied with decreased CCR6+ and higher CXCR3+ regulatory T cells (Tregs) frequency. These conditions result in exacerbated MOG35-55 peptide-induced EAE. Gal-8 eliminates activated Th17 but not Th1 cells by apoptosis and ameliorates EAE in C57BL/6 wild-type mice. ß-gal histochemistry reflecting the activity of the Gal-8 promoter revealed Gal-8 expression in a wide range of CNS regions, including high expression in the choroid-plexus. Accordingly, we detected Gal-8 in human cerebrospinal fluid, suggesting a role in the CNS immune-surveillance circuit. In addition, we show that MS patients generate function-blocking anti-Gal-8 antibodies with pathogenic potential. Such antibodies block cell adhesion and Gal-8-induced Th17 apoptosis. Furthermore, circulating anti-Gal-8 antibodies associate with relapsing-remitting MS (RRMS), and not with progressive MS phenotypes, predicting clinical disability at diagnosis within the first year of follow-up. Our results reveal that Gal-8 has an immunosuppressive protective role against autoimmune CNS inflammation, modulating the balance of Th17 and Th1 polarization and their respective Tregs. Such a role can be counteracted during RRMS by anti-Gal-8 antibodies, worsening disease prognosis. Even though anti-Gal-8 antibodies are not specific for MS, our results suggest that they could be a potential early severity biomarker in RRMS.


Asunto(s)
Autoanticuerpos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Galectinas/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Apoptosis/fisiología , Encéfalo/inmunología , Encéfalo/metabolismo , Adhesión Celular/fisiología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Galectinas/genética , Galectinas/metabolismo , Silenciador del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Pronóstico , Linfocitos T Reguladores/metabolismo , Células Th17/metabolismo
7.
Brain Behav Immun ; 37: 187-96, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24380849

RESUMEN

Aging is the main risk factor for Alzheimer's disease. Among other characteristics, it shows changes in inflammatory signaling that could affect the regulation of glial cell activation. We have shown that astrocytes prevent microglial cell cytotoxicity by mechanisms mediated by TGFß1. However, whereas TGFß1 is increased, glial cell activation persists in aging. To understand this apparent contradiction, we studied TGFß1-Smad3 signaling during aging and their effect on microglial cell function. TGFß1 induction and activation of Smad3 signaling in the hippocampus by inflammatory stimulation was greatly reduced in adult mice. We evaluated the effect of TGFß1-Smad3 pathway on the regulation of nitric oxide (NO) and reactive oxygen species (ROS) secretion, and phagocytosis of microglia from mice at different ages with and without in vivo treatment with lipopolysaccharide (LPS) to induce an inflammatory status. NO secretion was only induced on microglia from young mice exposed to LPS, and was potentiated by inflammatory preconditioning, whereas in adult mice the induction of ROS was predominant. TGFß1 modulated induction of NO and ROS production in young and adult microglia, respectively. Modulation was partially dependent on Smad3 pathway and was impaired by inflammatory preconditioning. Phagocytosis was induced by inflammation and TGFß1 only in microglia cultures from young mice. Induction by TGFß1 was also prevented by Smad3 inhibition. Our findings suggest that activation of the TGFß1-Smad3 pathway is impaired in aging. Age-related impairment of TGFß1-Smad3 can reduce protective activation while facilitating cytotoxic activation of microglia, potentiating microglia-mediated neurodegeneration.


Asunto(s)
Microglía/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Envejecimiento , Péptidos beta-Amiloides/metabolismo , Animales , Hipocampo/metabolismo , Inflamación/metabolismo , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Fagocitosis , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
Mol Cell Endocrinol ; 384(1-2): 71-82, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24447464

RESUMEN

11 beta-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) converts cortisone to cortisol in a NADPH dependent manner. Overexpression of 11ß-HSD1 in key metabolic tissues is related to the development of type 2 diabetes, obesity, hypertension and metabolic syndrome. Using crystal structures of human 11ß-HSD1 in complex with inhibitors as source of structural information, a combined ligand and structure-based virtual screening approach was implemented to identify novel 11ß-HSD1 inhibitors. A selected group of compounds was identified in silico and further evaluated in cell-based assays for cytotoxicity and 11ß-HSD1 mediated cortisol production inhibitory capacity. The expression of 11ß-HSD1 and 11ß-HSD2 in human LS14 adipocytes was assessed during differentiation. Biological evaluation of 39 compounds in adipocytes and steroids quantification by HPLC-MS/MS identify 4 compounds that exhibit 11ß-HSD1 mediated cortisol production inhibitory activity with potencies in the micromolar range. Two compounds showed to be selective for the 11ß-HSD1 reductase activity and over 11ß-HSD2 isoform, and thus represent novel leads for the development of more active derivatives with higher efficacies targeting intracellular cortisol levels in type 2 diabetes and metabolic syndrome.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/antagonistas & inhibidores , Adipocitos/efectos de los fármacos , Inhibidores Enzimáticos/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Adipocitos/citología , Adipocitos/enzimología , Diferenciación Celular , Línea Celular Tumoral , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Expresión Génica , Humanos , Hidrocortisona/antagonistas & inhibidores , Hidrocortisona/biosíntesis , Cinética , Ligandos , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Interfaz Usuario-Computador
9.
J Neurosci Res ; 90(10): 1970-80, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22715062

RESUMEN

Inflammatory cytokines and ß amyloid (Aß) induce activation of glial cells, leading to both protective and deleterious changes that are relevant for the pathogenesis of Alzheimer disease (AD). We have shown that astrocytes downregulate microglial cell cytotoxic activation through secretion of transforming growth factor-ß (TGFß1), and there is evidence that TGFß1 modifies Aß removal through the modulation of microglia. However, inflammatory activation of microglia is increased and Aß clearance is reduced in AD patients, regardless of the fact that TGFß1 is increased in their nervous system. We propose that changes in TGFß Smad3 signal transduction could modify the regulation mediated by TGFß1. Here we evaluated the participation of the TGFß Smad3 pathway in regulation of the expression pattern of scavenger receptors (SR) and activation of microglia through nitric oxide (NO·) secretion and phagocytosis of Aß. We found that TGFß1 increased SR-A by 2.4-fold and decreased SR-BI expression by 79% at 48 hr, whereas it did not change SR-MARCO or CD36 expression. In addition, we observed a 51% increase of Aß uptake and an 83% decrease of NO· production induced by lipopolysaccharide in microglial cell cultures. Increased expression of SR-A, phagocytosis, and downregulation of NO· by TGFß1 were prevented by the inhibition of the TGFß Smad3 pathway. Our results indicate that the modulation of microglial cell activation by TGFß1, leading to increased clearance of Aß and reduced cytotoxicity, is at least partially mediated by the Smad pathway.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Microglía/metabolismo , Proteína smad3/fisiología , Factor de Crecimiento Transformador beta1/farmacología , Animales , Western Blotting , Radicales Libres/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Inmunohistoquímica , Microglía/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Óxido Nítrico/metabolismo , Fagocitosis/efectos de los fármacos , Plásmidos/genética , Ratas , Ratas Sprague-Dawley , Receptores Depuradores/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína smad3/genética , Transfección , Tubulina (Proteína)/metabolismo
10.
Hypertension ; 59(1): 85-91, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22083159

RESUMEN

Familial hyperaldosteronism type I is caused by an unequal crossover of 11ß-hydroxylase (CYP11B1) and aldosterone synthase (CYP11B2) genes, giving rise to a chimeric CYP11B1/CYP11B2 gene (CG). We describe a family carrying a CG with high levels of free 18-hydroxycortisol but low prevalence of primary aldosteronism (PA) and an atypical CG inheritance pattern in a family of 4 generations with 16 adults and 13 children, we measured the arterial blood pressure, serum aldosterone, and plasma renin activity and then calculated the serum aldosterone:plasma renin activity ratio and urinary free 18-hydroxycortisol. We identified the CG by long-extension PCR and predicted its inheritance pattern. The CG was found in 24 of 29 subjects (10 children and 14 adults). In CG+ patients, hypertension and high 18-hydroxycortisol were prevalent (83% and 100%, respectively). High serum aldosterone:plasma renin activity ratio was more frequent in pediatric than adult patients (80% versus 36%; P<0.001). An inverse association between serum aldosterone:plasma renin activity ratio and age was observed (r=-0.48; P=0.018). Sequence analysis identified the CYP11B1/CYP11B2 crossover in a 50-bp region spanning intron 3 of CYP11B1 and exon 4 of CYP11B2. The CG segregation differs from an autosomal disease, showing 100% of CG penetrance in generations II and III. Statistical analysis suggests that inheritance pattern was not attributed to random segregation (P<0.001). In conclusion, we describe a family with an atypical CYP11B1/CYP11B2 gene inheritance pattern and variable phenotypic expression, where the majority of pediatric patients have primary aldosteronism. Most adults have normal aldosterone and renin levels, which could mask them as essential hypertensives.


Asunto(s)
Segregación Cromosómica/genética , Citocromo P-450 CYP11B2/genética , Hiperaldosteronismo/epidemiología , Hiperaldosteronismo/genética , Esteroide 11-beta-Hidroxilasa/genética , Adolescente , Adulto , Aldosterona/sangre , Chile/epidemiología , Puntos de Rotura del Cromosoma , Salud de la Familia , Femenino , Humanos , Hiperaldosteronismo/sangre , Masculino , Proteínas Mutantes Quiméricas/genética , Linaje , Prevalencia
11.
J Neurosci Methods ; 202(1): 65-9, 2011 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-21889955

RESUMEN

Microglial cells' phenotype and function change with aging. Since microglial cell impairments that are relevant for neurodegenerative diseases appear to be unique to aged individuals, it is important to assess function of aged microglia. However, most studies are done with microglia from neonates, mostly due to lack of reliable protocols to obtain microglia from adult animals. Here, we present a conditioned media-dependent culture system that promotes proliferation of adult microglia. We observed that inflammatory activation was increasingly oxidative in microglia from aged animals. Also, whereas phagocytosis of Aß by microglia from adult animals was more robust than that of microglia from neonates, the induction of phagocytosis by TGFß was abolished in aged animals. Our results show the importance of using adult animals cells for the study of neurodegenerative processes or other diseases associated with aging. The proposed culture method is inexpensive and cell yield allows for their assessment by functional bioassays and biochemistry.


Asunto(s)
Envejecimiento/patología , Técnicas de Cultivo de Célula/métodos , Microglía/patología , Enfermedades Neurodegenerativas/patología , Envejecimiento/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Medios de Cultivo Condicionados , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Estrés Oxidativo/fisiología , Fagocitosis/fisiología
12.
J Neurochem ; 112(5): 1099-114, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20002526

RESUMEN

Among multiple structural and functional brain changes, aging is accompanied by an increase of inflammatory signaling in the nervous system as well as a dysfunction of the immune system elsewhere. Although the long-held view that aging involves neurocognitive impairment is now dismissed, aging is a major risk factor for neurodegenerative diseases such as Alzheimer;s disease, Parkinson;s disease and Huntington's disease, among others. There are many age-related changes affecting the brain, contributing both to certain declining in function and increased frailty, which could singly and collectively affect neuronal viability and vulnerability. Among those changes, both inflammatory responses in aged brains and the altered regulation of toll like receptors, which appears to be relevant for understanding susceptibility to neurodegenerative processes, are linked to pathogenic mechanisms of several diseases. Here, we review how aging and pro-inflammatory environment could modulate microglial phenotype and its reactivity and contribute to the genesis of neurodegenerative processes. Data support our idea that age-related microglial cell changes, by inducing cytotoxicity in contrast to neuroprotection, could contribute to the onset of neurodegenerative changes. This view can have important implications for the development of new therapeutic approaches.


Asunto(s)
Envejecimiento , Encéfalo/patología , Microglía/patología , Enfermedades Neurodegenerativas/patología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Expresión Génica/fisiología , Humanos , Hierro/metabolismo , Microglía/metabolismo , Estrés Oxidativo/fisiología
13.
World J Gastroenterol ; 13(22): 3071-9, 2007 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-17589922

RESUMEN

AIM: To examine the in vivo phenotype associated with hepatic metastatic lymph node 64 (MLN64) over-expression. METHODS: Recombinant-adenovirus-mediated MLN64 gene transfer was used to overexpress MLN64 in the livers of C57BL/6 mice. We measured the effects of MLN64 overexpression on hepatic cholesterol content, bile flow, biliary lipid secretion and apoptosis markers. For in vitro studies cultured CHO cells with transient MLN64 overexpression were utilized and apoptosis by TUNEL assay was measured. RESULTS: Livers from Ad.MLN64-infected mice exhibited early onset of liver damage and apoptosis. This response correlated with increases in liver cholesterol content and biliary bile acid concentration, and impaired bile flow. We investigated whether liver MLN64 expression could be modulated in a murine model of hepatic injury. We found increased hepatic MLN64 mRNA and protein levels in mice with chenodeoxycholic acid-induced liver damage. In addition, cultured CHO cells with transient MLN64 overexpression showed increased apoptosis. CONCLUSION: In summary, hepatic MLN64 over-expression induced damage and apoptosis in murine livers and altered cholesterol metabolism. Further studies are required to elucidate the relevance of these findings under physiologic and disease conditions.


Asunto(s)
Apoptosis , Hepatopatías/metabolismo , Hepatopatías/patología , Fosfoproteínas/metabolismo , Adenoviridae/genética , Fosfatasa Alcalina/sangre , Animales , Células CHO , Línea Celular , Enfermedad Hepática Inducida por Sustancias y Drogas , Ácido Quenodesoxicólico , Colesterol/metabolismo , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Fenotipo , Fosfoproteínas/genética , Transfección
14.
J Lipid Res ; 48(4): 924-34, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17234604

RESUMEN

Peroxisomal proliferators, such as ciprofibrate, are used extensively as effective hypolipidemic drugs. The effects of these compounds on lipid metabolism require ligand binding activation of the peroxisome proliferator-activated receptor (PPAR) alpha subtype of nuclear receptors and involve transcriptional activation of the metabolic pathways involved in lipid oxidative metabolism, transport, and disposition. omega-Hydroxylated-eicosatrienoic acids (HEETs), products of the sequential metabolism of arachidonic acid (AA) by the cytochrome P450 CYP2C epoxygenase and CYP4A omega-hydroxylase gene subfamilies, have been identified as potent and high-affinity ligands of PPARalpha in vitro and as PPARalpha activators in transient transfection assays. Using isolated rat hepatocytes in culture, we demonstrate that specific inhibition of either the CYP2C epoxygenase or the CYP4A omega-hydroxylase abrogates ciprofibrate-induced peroxisomal proliferation, whereas inhibition of other eicosanoid-synthesizing pathways had no effect. Conversely, overexpression of the rat liver CYP2C11 epoxygenase leads to spontaneous peroxisomal proliferation, an effect that is reversed by a CYP inhibitor. Based on these results, we propose that HEETs may serve as endogenous PPARalpha ligands and that the P450 AA monooxygenases participate in ciprofibrate-induced peroxisomal proliferation and the activation of PPARalpha downstream targets.


Asunto(s)
Ácido Clofíbrico/análogos & derivados , Citocromo P-450 CYP4A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , PPAR alfa/metabolismo , Peroxisomas/fisiología , Animales , Ácido Araquidónico/metabolismo , Células Cultivadas , Ácido Clofíbrico/farmacología , Ácidos Fíbricos , Hepatocitos/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Ligandos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...