Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Am J Respir Crit Care Med ; 189(5): 593-601, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24471484

RESUMEN

RATIONALE: An increased cancer aggressiveness and mortality have been recently reported among patients with obstructive sleep apnea (OSA). Intermittent hypoxia (IH), a hallmark of OSA, enhances melanoma growth and metastasis in mice. OBJECTIVES: To assess whether OSA-related adverse cancer outcomes occur via IH-induced changes in host immune responses, namely tumor-associated macrophages (TAMs). MEASUREMENTS AND MAIN RESULTS: Lung epithelial TC1 cell tumors were 84% greater in mice subjected to IH for 28 days compared with room air (RA). In addition, TAMs in IH-exposed tumors exhibited reductions in M1 polarity with a shift toward M2 protumoral phenotype. Although TAMs from tumors harvested from RA-exposed mice increased TC1 migration and extravasation, TAMs from IH-exposed mice markedly enhanced such effects and also promoted proliferative rates and invasiveness of TC1 cells. Proliferative rates of melanoma (B16F10) and TC1 cells exposed to IH either in single culture or in coculture with macrophages (RAW 264.7) increased only when RAW 264.7 macrophages were concurrently present. CONCLUSIONS: Our findings support the notion that IH-induced alterations in TAMs participate in the adverse cancer outcomes reported in OSA.


Asunto(s)
Hipoxia/inmunología , Neoplasias Pulmonares/patología , Macrófagos/patología , Melanoma Experimental/patología , Apnea Obstructiva del Sueño/fisiopatología , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Hipoxia/etiología , Neoplasias Pulmonares/inmunología , Masculino , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Fenotipo , Apnea Obstructiva del Sueño/inmunología , Linfocitos T Citotóxicos/patología
2.
J Neurosci ; 31(28): 10241-8, 2011 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-21753001

RESUMEN

Stroke is not only more prevalent but is also associated with more severe adverse functional outcomes among patients with sleep apnea. Monocarboxylate transporters (MCT) are important regulators of cellular bioenergetics, have been implicated in brain susceptibility to acute severe hypoxia (ASH), and could underlie the unfavorable prognosis of cerebrovascular accidents in sleep apnea patients. Rodents were exposed to either intermittent hypoxia (IH) during sleep, a characteristic feature of sleep apnea, or to sustained hypoxia (SH), and expression of MCT1 and MCT2 was assessed. In addition, the functional recovery to middle cerebral artery occlusion (MCAO) in rats and hMCT2 transgenic mice and of hippocampal slices subjected to ASH was assessed, as well as the effects of MCT blocker and MCT2 antisense oligonucleotides and siRNAs. IH, but not SH, induced significant reductions in MCT2 expression over time at both the mRNA and protein levels and in the functional recovery of hippocampal slices subjected to ASH. Similarly, MCAO-induced infarcts were significantly greater in IH-exposed rats and mice, and overexpression of hMCT2 in mice markedly attenuated the adverse effects of IH. Exogenous pyruvate treatment reduced infarct volumes in normoxic rats but not in IH-exposed rats. Administration of the MCT2 blocker 4CN, but not the MCT1 antagonist p-chloromercuribenzene sulfonate, increased infarct size. Thus, prolonged exposures to IH mimicking sleep apnea are associated with increased CNS vulnerability to ischemia that is mediated, at least in part, by concomitant decreases in the expression and function of MCT2. Efforts to develop agonists of MCT2 should provide opportunities to ameliorate the overall outcome of stroke.


Asunto(s)
Hipocampo/metabolismo , Hipoxia/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Síndromes de la Apnea del Sueño/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transportadores de Ácidos Monocarboxílicos/genética , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Síndromes de la Apnea del Sueño/complicaciones , Síndromes de la Apnea del Sueño/fisiopatología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/fisiopatología
3.
J Neuroinflammation ; 9: 91, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22578011

RESUMEN

BACKGROUND: Sleepiness and cognitive dysfunction are recognized as prominent consequences of sleep deprivation. Experimentally induced short-term sleep fragmentation, even in the absence of any reductions in total sleep duration, will lead to the emergence of excessive daytime sleepiness and cognitive impairments in humans. Tumor necrosis factor (TNF)-α has important regulatory effects on sleep, and seems to play a role in the occurrence of excessive daytime sleepiness in children who have disrupted sleep as a result of obstructive sleep apnea, a condition associated with prominent sleep fragmentation. The aim of this study was to examine role of the TNF-α pathway after long-term sleep fragmentation in mice. METHODS: The effect of chronic sleep fragmentation during the sleep-predominant period on sleep architecture, sleep latency, cognitive function, behavior, and inflammatory markers was assessed in C57BL/6 J and in mice lacking the TNF-α receptor (double knockout mice). In addition, we also assessed the above parameters in C57BL/6 J mice after injection of a TNF-α neutralizing antibody. RESULTS: Mice subjected to chronic sleep fragmentation had preserved sleep duration, sleep state distribution, and cumulative delta frequency power, but also exhibited excessive sleepiness, altered cognitive abilities and mood correlates, reduced cyclic AMP response element-binding protein phosphorylation and transcriptional activity, and increased phosphodiesterase-4 expression, in the absence of AMP kinase-α phosphorylation and ATP changes. Selective increases in cortical expression of TNF-α primarily circumscribed to neurons emerged. Consequently, sleepiness and cognitive dysfunction were absent in TNF-α double receptor knockout mice subjected to sleep fragmentation, and similarly, treatment with a TNF-α neutralizing antibody abrogated sleep fragmentation-induced learning deficits and increases in sleep propensity. CONCLUSIONS: Taken together, our findings show that recurrent arousals during sleep, as happens during sleep apnea, induce excessive sleepiness via activation of inflammatory mechanisms, and more specifically TNF-α-dependent pathways, despite preserved sleep duration.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Transducción de Señal/fisiología , Privación de Sueño/metabolismo , Fases del Sueño/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Nivel de Alerta/genética , Nivel de Alerta/fisiología , Encéfalo/metabolismo , Encéfalo/fisiología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/psicología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vías Nerviosas/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Recurrencia , Transducción de Señal/genética , Privación de Sueño/genética , Privación de Sueño/psicología , Fases del Sueño/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética
4.
BMC Neurosci ; 13: 77, 2012 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-22759774

RESUMEN

BACKGROUND: In rodents, exposure to intermittent hypoxia (IH), a hallmark of obstructive sleep apnea (OSA), is associated with neurobehavioral impairments, increased apoptosis in the hippocampus and cortex, as well as increased oxidant stress and inflammation. Such findings are markedly attenuated in rodents exposed to sustained hypoxia 9SH) of similar magnitude. The hypoxia-sensitive gene erythropoietin (EPO) has emerged as a major endogenous neuroprotectant, and could be involved in IH-induced neuronal dysfunction. METHODS AND RESULTS: IH induced only transiently increased expression of EPO mRNA in hippocampus, which was continued in (SH)-exposed mice. IH, but not SH, adversely affected two forms of spatial learning in the water maze, and increased markers of oxidative stress. However, on a standard place training task, mice treated with exogenously administered EPO displayed normal learning, and were protected from the spatial learning deficits observed in vehicle-treated (C) littermates exposed to IH. Moreover, anxiety levels were increased in IH as compared to normoxia, while no changes in anxiety emerged in EPO-treated mice. Additionally, C mice, but not EPO-treated IH-exposed mice had significantly elevated levels of NADPH oxidase expression, as well as increased MDA and 8-OHDG levels in cortical and hippocampal lysates. CONCLUSIONS: The oxidative stress responses and neurobehavioral impairments induced by IH during sleep are mediated, at least in part, by imbalances between EPO expression and increased NADPH oxidase activity, and thus pharmacological agents targeting EPO expression in CNS may provide a therapeutic strategy in sleep-disordered breathing.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Eritropoyetina/uso terapéutico , Hipoxia/complicaciones , Síndromes de la Apnea del Sueño/complicaciones , 8-Hidroxi-2'-Desoxicoguanosina , Análisis de Varianza , Animales , Células Cultivadas , Corteza Cerebral/citología , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Embrión de Mamíferos , Eritropoyetina/genética , Eritropoyetina/metabolismo , Reacción de Fuga/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Inyecciones Intraperitoneales , Peroxidación de Lípido/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Natación/psicología , Factores de Tiempo
5.
Am J Respir Crit Care Med ; 184(11): 1305-12, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21868506

RESUMEN

RATIONALE: Sleep fragmentation (SF) is one of the major characteristics of sleep apnea, and has been implicated in its morbid consequences, which encompass excessive daytime sleepiness and neurocognitive impairments. We hypothesized that absence of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity is neuroprotective in SF-induced cognitive impairments. OBJECTIVES: To examine whether increased NADPH oxidase activity may play a role in SF-induced central nervous system dysfunction. METHODS: The effect of chronic SF during the sleep-predominant period on sleep architecture, sleep latency, spatial memory, and oxidative stress parameters was assessed in mice lacking NADPH oxidase activity (gp91phox-(/Y)) and wild-type littermates. MEASUREMENTS AND MAIN RESULTS: SF for 15 days was not associated with differences in sleep duration, sleep state distribution, or sleep latency in both gp91phox-(/Y) and control mice. However, on a standard place training task, gp91phox-(/Y) mice displayed normal learning and were protected from the spatial learning deficits observed in wild-type littermates exposed to SF. Moreover, anxiety levels were increased in wild-type mice exposed to SF, whereas no changes emerged in gp91phox-(/Y) mice. Additionally, wild-type mice, but not gp91phox-(/Y) mice, had significantly elevated NADPH oxidase gene expression and activity, and in malondialdehyde and 8-oxo-2'-deoxyguanosine levels in cortical and hippocampal lysates after SF exposures. CONCLUSIONS: This work substantiates an important role for NADPH oxidase in hippocampal memory impairments induced by SF, modeling sleep apnea. Targeting NADPH oxidase, therefore, is expected to minimize hippocampal impairments from both intermittent hypoxia and SF associated with the disease.


Asunto(s)
Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/metabolismo , NADPH Oxidasas/metabolismo , Privación de Sueño/complicaciones , Privación de Sueño/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Análisis de Varianza , Animales , Conducta Animal , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animales de Enfermedad , Peroxidación de Lípido , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Estrés Oxidativo
6.
Am J Physiol Cell Physiol ; 299(2): C497-505, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20538765

RESUMEN

Glutathione transport into mitochondria is mediated by oxoglutarate (OGC) and dicarboxylate carrier (DIC) in the kidney and liver. However, transport mechanisms in brain mitochondria are unknown. We found that both carriers were expressed in the brain. Using cortical mitochondria incubated with physiological levels of glutathione, we found that butylmalonate, a DIC inhibitor, reduced mitochondrial glutathione to levels similar to those seen in mitochondria incubated without extramitochondrial glutathione (59% of control). In contrast, phenylsuccinate, an OGC inhibitor, had no effect (97% of control). Additional experiments with DIC and OGC short hairpin RNA in neuronal-like PC12 cells resulted in similar findings. Significantly, DIC inhibition resulted in increased reactive oxygen species (ROS) content in and H(2)O(2) release from mitochondria. It also led to decreased membrane potential, increased basal respiration rates, and decreased phosphorus-to-oxygen (P/O) ratios, especially when electron transport was initiated from complex I. Accordingly, we found that DIC inhibition impaired complex I activity, but not those for complexes II and III. This impairment was not associated with dislodgment of complex subunits. These results suggest that DIC is the main glutathione transporter in cortical mitochondria and that DIC-mediated glutathione transport is essential for these mitochondria to maintain ROS homeostasis and normal respiratory functions.


Asunto(s)
Encéfalo/metabolismo , Transportadores de Ácidos Dicarboxílicos/fisiología , Glutatión/metabolismo , Homeostasis/fisiología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Animales Recién Nacidos , Transporte Biológico/fisiología , Respiración de la Célula/fisiología , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Células PC12 , Ratas , Ratas Sprague-Dawley
8.
Am J Respir Cell Mol Biol ; 40(3): 332-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18776132

RESUMEN

Whole-body hypoxic preconditioning (WHPC) prolongs survival of mice exposed to severe hypoxia by attenuating pulmonary edema and preserving gas exchange. However, the cellular and molecular mechanism(s) of this protection remains unclear. The objective of this study was to identify the cellular target(s) of WHPC in the lung. Conscious mice were exposed to hypoxia (7% O(2)) for 6 hours with or without pretreatment of WHPC ([8% O(2)] x 10 min/[21% O(2)] x 10 min; 6 cycles). Hypoxia caused severe lung injury, as shown by the development of high-permeability-type pulmonary edema and the release of lactate dehydrogenase and creatine kinase into the airspace and the circulation. All these signs of hypoxic lung injury were significantly attenuated by WHPC. Hypoxia also caused a remarkable release of type I cell markers (caveolin-2 and receptor for advanced glycation end products) in lung lavage that was almost completely abolished by WHPC. Conversely, hypoxia-induced release of type II cell markers (surfactant-associated proteins A and D) was only marginal, and was unaffected by WHPC. Electron microscopic analysis demonstrated considerable hypoxic damage in alveolar type I cells and vascular endothelial cells. Notably, WHPC completely eliminated hypoxic damage in the former and alleviated it in the latter. Type II cells appeared normal. Furthermore, WHPC up-regulated protein expression of cytoprotective genes in the lung, such as heat shock proteins and manganese superoxide dismutase. Thus, WHPC attenuates hypoxic lung injury through protection of cells constituting the respiratory membrane, especially hypoxia-vulnerable type I epithelial cells. This beneficial effect may involve up-regulation of cytoprotective genes.


Asunto(s)
Células Epiteliales/fisiología , Hipoxia/metabolismo , Lesión Pulmonar/patología , Pulmón/citología , Pulmón/fisiología , Animales , Biomarcadores/metabolismo , Líquido del Lavado Bronquioalveolar/citología , Citoprotección/genética , Células Epiteliales/ultraestructura , Hipoxia/patología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de von Willebrand/inmunología , Factor de von Willebrand/metabolismo
9.
Physiol Genomics ; 32(1): 95-104, 2007 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-17911380

RESUMEN

Expression patterns of monocarboxylate transporter 2 (MCT2) display mRNA diversity in a tissue-specific fashion. We cloned and characterized multiple mct2 5'-cDNA ends from the mouse and determined the structural organization of the mct2 gene. We found that transcription of this gene was initiated from five independent genomic regions that spanned >80 kb on chromosome 10, resulting in five unique exon 1 variants (exons 1a, 1b, 1c, 1d, and 1e) that were then spliced to the common exon 2. Alternative splicing of four internal exons (exons AS1, AS2, AS3, and exon 3) greatly increased the complexity of mRNA diversity. While exon 1c was relatively commonly used for transcription initiation in various tissues, other exon 1 variants were used in a tissue-specific fashion, especially exons 1b and 1d that were used exclusively for testis-specific expression. Sequence analysis of 5'-flanking regions upstream of exons 1a, 1b, and 1c revealed the presence of numerous potential binding sites for ubiquitous transcription factors in all three regions and for transcription factors implicated in testis-specific or hypoxia-induced gene expression in the 1b region. Transient transfection assays demonstrated that each of the three regions contained a functional promoter and that the in vitro, cell type-specific activities of these promoters were consistent with the tissue-specific expression pattern of the mct2 gene in vivo. These results indicate that tissue-specific expression of the mct2 gene is controlled by multiple alternative promoters and that both alternative promoter usage and alternative splicing contribute to the remarkable mRNA diversity of the gene.


Asunto(s)
Empalme Alternativo , Transportadores de Ácidos Monocarboxílicos/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , Animales , Secuencia de Bases , Exones , Amplificación de Genes , Regulación de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Circ Res ; 95(1): 84-91, 2004 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-15166094

RESUMEN

The infarct-sparing effect of the late phase of ischemic preconditioning (late PC) lasts for 72 hours. Upregulation of both cyclooxygenase-2 (COX-2) and inducible NO synthase (iNOS) has been shown to be essential to the protection in the initial stage of late PC (24 hours after PC); however, the mechanisms underlying the protection in the final stage of late PC (48 to 72 hours after PC) are unknown. Conscious rabbits were preconditioned with six cycles of 4-minute coronary occlusion/4-minute reperfusion. At 72 hours after PC, powerful protection against infarction was associated with increased myocardial levels of COX-2 mRNA, protein, and cardioprotective prostaglandins (PGI2 and PGE2). The COX-2-selective inhibitor NS-398 completely blocked the protection. Surprisingly, iNOS expression was not increased at 72 hours; instead, upregulation of neuronal NO synthase (nNOS) was evident at both the mRNA (+266+/-20%, P<0.005) and the protein levels (+195+/-66%, P<0.005), which was accompanied by an increase in myocardial nitrite/nitrate (+20+/-4%, P<0.05). The nNOS-selective inhibitors N-propyl-l-arginine or S-ethyl N-[4-(trifluoromethyl)phenyl]isothiourea completely blocked the protection of late PC at 72 hours, whereas the iNOS-selective inhibitor S-methylisothiourea had no effect. In line with these findings, the disappearance of protection at 120 hours after PC was associated with the return of nNOS mRNA, protein, and activity to control levels. Although expression of COX-2 protein was still elevated at 120 hours, only a marginal increase in PGI2 and PGE2 levels was detected. In contrast to 72 hour after PC, nNOS was not upregulated at 24 hour after PC. We conclude that (1) the cardioprotection observed in the final stage of late PC (72 hour) is mediated by nNOS, not by iNOS, in concert with COX-2, and (2) nNOS-derived NO is required to drive COX-2 activity. These data identify, for the first time, a cardioprotective role of nNOS and demonstrate, surprisingly, that the mechanism of late PC differs at 72 hours (nNOS) versus 24 hours (iNOS).


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Isoenzimas/metabolismo , Infarto del Miocardio/prevención & control , Óxido Nítrico Sintasa/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Secuencia de Aminoácidos , Animales , Ciclooxigenasa 2 , Isoenzimas/fisiología , Datos de Secuencia Molecular , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Óxido Nítrico Sintasa/química , Óxido Nítrico Sintasa/fisiología , Óxido Nítrico Sintasa de Tipo I , Prostaglandina-Endoperóxido Sintasas/fisiología , Conejos , Factores de Tiempo , Regulación hacia Arriba
11.
FASEB J ; 17(12): 1709-11, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12958184

RESUMEN

In neurons, hypoxia activates intracellular death-related pathways, yet the antiapoptotic mechanisms triggered by hypoxia remain unclear. In RN46A neuronal cells, minimum media growth conditions induced cell death as early as 12 h after the cells were placed in these conditions (i.e., after removal of B-27 supplement). However, apoptosis occurred in hypoxia (1% O2) only after 48 h, and in fact hypoxia reduced the apoptosis associated with trophic factor withdrawal. Furthermore, hypoxia induced time-dependent increases in expression of platelet-derived growth factor (PDGF) B mRNA and protein, as well as PDGF-beta receptor phosphorylation. Although exogenous PDGF-BB induced only transient Akt activation, hypoxia triggered persistent activation of Akt for up to 24 h. Inhibition of phosphatidylinositol 3-kinase (PI3K) or of PDGF-beta receptor phosphorylation abrogated both hypoxia-induced and exogenous PDGF-BB-induced Akt phosphorylation, and it completely abolished hypoxia-induced protection from media supplement deprivation, which suggests that the long-lasting activation of Akt during hypoxia and the prosurvival induction were due to endogenously generated PDGF-BB. Furthermore, these inhibitors decreased hypoxia-inducible factor 1alpha (HIF-1alpha) DNA binding, which suggests that the PDGF/PDGF-beta receptor/Akt pathway induces downstream HIF-1alpha gene transcription. We conclude that in RN46A neuronal cells, hypoxia activates an autocrine-paracrine antiapoptotic mechanism that involves up-regulation of PDGF-B and PDGF-beta receptor-dependent activation of the PI3K/Akt signaling pathway to induce downstream transcription of survival genes.


Asunto(s)
Comunicación Autocrina , Neuronas/metabolismo , Comunicación Paracrina , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-sis/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Apoptosis , Hipoxia de la Célula , Línea Celular , Supervivencia Celular , Medios de Cultivo , Subunidad alfa del Factor 1 Inducible por Hipoxia , Cinética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Neuronas/enzimología , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Factores de Transcripción/biosíntesis
12.
Endocrinology ; 156(2): 437-43, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25406018

RESUMEN

Chronic intermittent hypoxia during sleep (IH), as occurs in sleep apnea, promotes systemic insulin resistance. Resveratrol (Resv) has been reported to ameliorate high-fat diet-induced obesity, inflammation, and insulin resistance. To examine the effect of Resv on IH-induced metabolic dysfunction, male mice were subjected to IH or room air conditions for 8 weeks and treated with either Resv or vehicle (Veh). Fasting plasma levels of glucose, insulin, and leptin were obtained, homeostatic model assessment of insulin resistance index levels were calculated, and insulin sensitivity tests (phosphorylated AKT [also known as protein kinase B]/total AKT) were performed in 2 visceral white adipose tissue (VWAT) depots (epididymal [Epi] and mesenteric [Mes]) along with flow cytometry assessments for VWAT macrophages and phenotypes (M1 and M2). IH-Veh and IH-Resv mice showed initial reductions in food intake with later recovery, with resultant lower body weights after 8 weeks but with IH-Resv showing better increases in body weight vs IH-Veh. IH-Veh and IH-Resv mice exhibited lower fasting glucose levels, but only IH-Veh had increased homeostatic model assessment of insulin resistance index vs all 3 other groups. Leptin levels were preserved in IH-Veh but were significantly lower in IH-Resv. Reduced VWAT phosphorylated-AKT/AKT responses to insulin emerged in both Mes and Epi in IH-Veh but normalized in IH-Resv. Increases total macrophage counts and in M1 to M2 ratios occurred in IH-Veh Mes and Epi compared all other 3 groups. Thus, Resv ameliorates food intake and weight gain during IH exposures and markedly attenuates VWAT inflammation and insulin resistance, thereby providing a potentially useful adjunctive therapy for metabolic morbidity in the context of sleep apnea.


Asunto(s)
Fármacos Antiobesidad/farmacología , Hipoxia/inmunología , Resistencia a la Insulina , Grasa Intraabdominal/inmunología , Macrófagos/efectos de los fármacos , Estilbenos/farmacología , Animales , Evaluación Preclínica de Medicamentos , Ingestión de Alimentos , Insulina/sangre , Leptina/sangre , Masculino , Ratones Endogámicos C57BL , Distribución Aleatoria , Resveratrol , Aumento de Peso
13.
Oncoimmunology ; 4(2): e976057, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25949873

RESUMEN

The molecular mechanisms underlying how sleep fragmentation (SF) influences cancer growth and progression remain largely elusive. Here, we present evidence that SF reduced ROS production by downregulating gp91phox expression and activity in TC1 cell tumor associated macrophages (TAMs), while genetic ablation of phagocytic Nox2 activity increased tumor cell proliferation, motility, invasion, and extravasation in vitro. Importantly, the in vivo studies using immunocompetent syngeneic murine tumor models suggested that Nox2 deficiency mimics SF-induced TAMs infiltration and subsequent tumor growth and invasion. Taken together, these studies reveal that perturbed sleep could adversely affect innate immunity within the tumor by altering Nox2 expression and activity, and indicate that selective potentiation of Nox2 activity may present a novel therapeutic strategy in the treatment of cancer.

14.
Cancer Lett ; 361(2): 233-9, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25779675

RESUMEN

Intermittent hypoxia (IH)-induces alterations in tumor-associated macrophages (TAMs) that are associated with adverse cancer outcomes, as reported in patients suffering from sleep apnea. Adipose tissues (AT) and bone-marrow (BM)-derived cells are the inferred sources of macrophages infiltrating malignant tumors. Here, the sources of TAMs and the phenotypic changes induced by IH in the ipsilateral and contralateral AT were investigated by using a syngeneic murine solid tumor model (TC1). C57/B6 male mice were exposed to either IH or room air (RA) for 6 weeks, with TC1 cells being inoculated in the 2nd week. Macrophage content, phenotype and tissue origin were assessed in tumors, and ipsilateral and contralateral AT. IH induced a ~2.2-fold increase in TAM tumor infiltration. However, differential responses in the tumor ipsilateral and contralateral AT emerged: IH increased infiltration of preferentially M1 macrophages in contralateral AT, while reductions in macrophages emerged in ipsilateral AT and primarily consisted of the M2 phenotype. These changes were accompanied by reciprocal increases in resident and BM-derived TAMs in the tumor. IH-induced phenotypic alterations in AT macrophages surrounding the tumor and their increased infiltration within the tumor may contribute to the accelerated tumor progression associated with IH.


Asunto(s)
Tejido Adiposo/patología , Neoplasias Pulmonares/patología , Macrófagos/patología , Apnea Obstructiva del Sueño/patología , Microambiente Tumoral/fisiología , Tejido Adiposo/metabolismo , Animales , Hipoxia de la Célula/fisiología , Modelos Animales de Enfermedad , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Apnea Obstructiva del Sueño/metabolismo , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
15.
J Appl Physiol (1985) ; 96(1): 392-7, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14660501

RESUMEN

Survival in severe hypoxia such as occurs in high altitude requires previous acclimatization, which is acquired over a period of days to weeks. It was unknown whether intrinsic mechanisms existed that could be rapidly induced and could exert immediate protection on unacclimatized individuals against acute hypoxia. We found that mice pretreated with whole-body hypoxic preconditioning (WHPC, 6 cycles of 10-min hypoxia-10-min normoxia) survived significantly longer than control animals when exposed to lethal hypoxia (5% O2, survival time of 33.2 +/- 6.1 min vs. controls at 13.8 +/- 1.2 min, n = 10, P < 0.005). This protective mechanism became operative shortly after WHPC and remained effective for at least 8 h. Accordingly, mice subjected to WHPC demonstrated improved gas exchange when exposed to sublethal hypoxia (7% O2, arterial blood Po2 of 49.9 +/- 4.2 vs. controls at 39.7 +/- 3.6 Torr, n = 6, P < 0.05), reduced formation of pulmonary edema (increase in lung water of 0.491 +/- 0.111 vs. controls at 0.894 +/- 0.113 mg/mg dry tissue, n = 10, P < 0.02), and decreased pulmonary vascular permeability (lung lavage albumin of 7.63 +/- 0.63 vs. controls at 18.24 +/- 3.39 mg/dl, n = 6-10, P < 0.025). In addition, the severity of cerebral edema caused by exposure to sublethal hypoxia was also reduced after WHPC (increase in brain water of 0.254 +/- 0.052 vs. controls at 0.491 +/- 0.034 mg/mg dry tissue, n = 10, P < 0.01). Thus WHPC protects unacclimatized mice against acute and otherwise lethal hypoxia, and this protection involves preservation of vital organ functions.


Asunto(s)
Hipoxia/fisiopatología , Precondicionamiento Isquémico , Pulmón/fisiología , Edema Pulmonar/prevención & control , Enfermedad Aguda , Animales , Encéfalo/metabolismo , Dióxido de Carbono/sangre , Agua Pulmonar Extravascular/metabolismo , Hipoxia/mortalidad , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/sangre , Edema Pulmonar/mortalidad , Edema Pulmonar/fisiopatología , Intercambio Gaseoso Pulmonar/fisiología , Tasa de Supervivencia
16.
Sleep ; 37(5): 999-1009, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24790279

RESUMEN

BACKGROUND: Chronic sleep fragmentation (SF) without sleep curtailment induces increased adiposity. However, it remains unclear whether mobilization, proliferation, and differentiation of adipocyte progenitors (APs) occurs in visceral white adipose tissue (VWAT), and whether nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2) activity plays a role. METHODS: Changes in VWAT depot cell size and AP proliferation were assessed in wild-type and Nox2 null male mice exposed to SF and control sleep (SC). To assess mobilization, proliferation, and differentiation of bone marrow mesenchymal stem cells (BM-MSC), Sca-1+ bone marrow progenitors were isolated from GFP+ or RFP+ mice, and injected intravenously to adult male mice (C57BL/6) previously exposed to SF or SC. RESULTS: In comparison with SC, SF was associated with increased weight accrual at 3 w and thereafter, larger subcutaneous and visceral fat depots, and overall adipocyte size at 8 w. Increased global AP numbers in VWAT along with enhanced AP BrDU labeling in vitro and in vivo emerged in SF. Systemic injections of GFP+ BM-MSC resulted in increased AP in VWAT, as well as in enhanced differentiation into adipocytes in SF-exposed mice. No differences occurred between SF and SC in Nox2 null mice for any of these measurements. CONCLUSIONS: Chronic sleep fragmentation (SF) induces obesity in mice and increased proliferation and differentiation of adipocyte progenitors (AP) in visceral white adipose tissue (VWAT) that are mediated by increased Nox2 activity. In addition, enhanced migration of bone marrow mesenchymal stem cells from the systemic circulation into VWAT, along with AP differentiation, proliferation, and adipocyte formation occur in SF-exposed wild-type but not in oxidase 2 (Nox2) null mice. Thus, Nox2 may provide a therapeutic target to prevent obesity in the context of sleep disorders.


Asunto(s)
Adipocitos/citología , Tejido Adiposo Blanco/citología , Diferenciación Celular , Grasa Intraabdominal/citología , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Privación de Sueño/enzimología , Células Madre/citología , Adipocitos/enzimología , Tejido Adiposo Blanco/enzimología , Adiposidad , Animales , Movimiento Celular , Grasa Intraabdominal/enzimología , Masculino , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADP/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasas/deficiencia , Obesidad/complicaciones , Obesidad/enzimología , Obesidad/genética , Obesidad/patología , Privación de Sueño/complicaciones , Privación de Sueño/genética , Privación de Sueño/patología , Células Madre/enzimología
17.
Sleep ; 37(11): 1817-24, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25364077

RESUMEN

STUDY OBJECTIVES: Sleep fragmentation (SF) is a common occurrence and constitutes a major characteristic of obstructive sleep apnea (OSA). SF has been implicated in multiple OSA-related morbidities, but it is unclear whether SF underlies any of the cardiovascular morbidities of OSA. We hypothesized that long-term SF exposures may lead to endothelial dysfunction and altered vessel wall structure. METHODS AND RESULTS: Adult male C57BL/6J mice were fed normal chow and exposed to daylight SF or control sleep (CTL) for 20 weeks. Telemetric blood pressure and endothelial function were assessed weekly using a modified laser-Doppler hyperemic test. Atherosclerotic plaques, elastic fiber disruption, lumen area, wall thickness, foam cells, and macrophage recruitment, as well as expression of senescence-associated markers were examined in excised aortas. Increased latencies to reach baseline perfusion levels during the post-occlusive period emerged in SF mice with increased systemic BP values starting at 8 weeks of SF and persisting thereafter. No obvious atherosclerotic plaques emerged, but marked elastic fiber disruption and fiber disorganization were apparent in SF-exposed mice, along with increases in the number of foam cells and macrophages in the aorta wall. Senescence markers showed reduced TERT and cyclin A and increased p16INK4a expression, with higher IL-6 plasma levels in SF-exposed mice. CONCLUSIONS: Long-term sleep fragmentation induces vascular endothelial dysfunction and mild blood pressure increases. Sleep fragmentation also leads to morphologic vessel changes characterized by elastic fiber disruption and disorganization, increased recruitment of inflammatory cells, and altered expression of senescence markers, thereby supporting a role for sleep fragmentation in the cardiovascular morbidity of OSA.


Asunto(s)
Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Apnea Obstructiva del Sueño/complicaciones , Privación de Sueño/patología , Privación de Sueño/fisiopatología , Animales , Aorta/inmunología , Aorta/patología , Biomarcadores/sangre , Presión Sanguínea , Senescencia Celular , Quimiocina CXCL1/sangre , Ingestión de Alimentos , Tejido Elástico/patología , Frecuencia Cardíaca , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Interleucina-6/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Placa Aterosclerótica/patología , Apnea Obstructiva del Sueño/sangre , Apnea Obstructiva del Sueño/patología , Apnea Obstructiva del Sueño/fisiopatología , Privación de Sueño/sangre , Privación de Sueño/complicaciones
18.
Obesity (Silver Spring) ; 22(3): 758-62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24039209

RESUMEN

OBJECTIVES: Short sleep confers a higher risk of obesity in humans. Restricted sleep increases appetite, promotes higher calorie intake from fat and carbohydrate sources, and induces insulin resistance. However, the effects of fragmented sleep (SF), such as occurs in sleep apnea, on body weight, metabolic rates, and adipose tissue distribution are unknown. METHODS: C57BL/6 mice were exposed to SF for 8 weeks. Their body weight, food consumption, and metabolic expenditure were monitored over time, and their plasma leptin levels measured after exposure to SF for 1 day as well as for 2 weeks. In addition, adipose tissue distribution was assessed at the end of the SF exposure using MRI techniques. RESULTS: Chronic SF-induced obesogenic behaviors and increased weight gain in mice by promoting increased caloric intake without changing caloric expenditure. Plasma leptin levels initially decreased and subsequently increased. Furthermore, increases in both visceral and subcutaneous adipose tissue volumes occurred. CONCLUSIONS: These results suggest that SF, a frequent occurrence in many disorders and more specifically in sleep apnea, is a potent inducer of obesity via activation of obesogenic behaviors and possibly leptin resistance, in the absence of global changes in energy expenditure.


Asunto(s)
Obesidad/fisiopatología , Privación de Sueño/fisiopatología , Animales , Distribución de la Grasa Corporal , Calorimetría Indirecta , Modelos Animales de Enfermedad , Ingestión de Energía , Homeostasis , Grasa Intraabdominal/metabolismo , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Privación de Sueño/complicaciones , Grasa Subcutánea/metabolismo , Aumento de Peso
19.
Cancer Res ; 74(5): 1329-37, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24448240

RESUMEN

Sleep fragmentation (SF) is a highly prevalent condition and a hallmark of sleep apnea, a condition that has been associated with increased cancer incidence and mortality. In this study, we examined the hypothesis that sleep fragmentation promotes tumor growth and progression through proinflammatory TLR4 signaling. In the design, we compared mice that were exposed to sleep fragmentation one week before engraftment of syngeneic TC1 or LL3 tumor cells and tumor analysis four weeks later. We also compared host contributions through the use of mice genetically deficient in TLR4 or its effector molecules MYD88 or TRIF. We found that sleep fragmentation enhanced tumor size and weight compared with control mice. Increased invasiveness was apparent in sleep fragmentation tumors, which penetrated the tumor capsule into surrounding tissues, including adjacent muscle. Tumor-associated macrophages (TAM) were more numerous in sleep fragmentation tumors, where they were distributed in a relatively closer proximity to the tumor capsule compared with control mice. Although tumors were generally smaller in both MYD88(-/-) and TRIF(-/-) hosts, the more aggressive features produced by sleep fragmentation persisted. In contrast, these more aggressive features produced by sleep fragmentation were abolished completely in TLR4(-/-) mice. Our findings offer mechanistic insights into how sleep perturbations can accelerate tumor growth and invasiveness through TAM recruitment and TLR4 signaling pathways.


Asunto(s)
Neoplasias/genética , Neoplasias/patología , Transducción de Señal/genética , Privación de Sueño/complicaciones , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Progresión de la Enfermedad , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Neoplasias/metabolismo
20.
Compr Physiol ; 2(3): 1767-77, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23723023

RESUMEN

Intermittent hypoxia (IH) is a frequent occurrence in clinical settings. In the last decades, evidence has emerged implicating the gas exchange alterations and sleep disruption associated with those disorders in the high prevalence of cognitive and behavioral deficits afflicting these patients. In an effort to better characterize the role of IH, and to identify potential mechanisms of IH-induced central nervous system (CNS) dysfunction, a large number of rodent models have been recently developed. The cumulative evidence confirms that IH indeed induces a heterotopic pattern of injury in the brain, particularly affecting cortical, subcortical, and hippocampal regions, ultimately leading to neuronal apoptosis and activation of microglia. These IH-induced deleterious processes exhibit substantial variability across the lifespan, are under substantial modulatory influences of diet, physical or intellectual activity, and genetic factors, and preferentially recruit oxidative stress and inflammatory pathways.


Asunto(s)
Enfermedades del Sistema Nervioso Central/etiología , Hipoxia/fisiopatología , Enfermedades del Sistema Nervioso Central/fisiopatología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/fisiopatología , Humanos , Hipoxia/complicaciones , Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA