Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 94(9)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32051269

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causal agent for Kaposi's sarcoma (KS), the most common malignancy in people living with human immunodeficiency virus (HIV)/AIDS. The oral cavity is a major route for KSHV infection and transmission. However, how KSHV breaches the oral epithelial barrier for spreading to the body is not clear. Here, we show that exosomes purified from either the saliva of HIV-positive individuals or the culture supernatants of HIV-1-infected T-cell lines promote KSHV infectivity in immortalized and primary human oral epithelial cells. HIV-associated saliva exosomes contain the HIV trans-activation response element (TAR), Tat, and Nef RNAs but do not express Tat and Nef proteins. The TAR RNA in HIV-associated exosomes contributes to enhancing KSHV infectivity through the epidermal growth factor receptor (EGFR). An inhibitory aptamer against TAR RNA reduces KSHV infection facilitated by the synthetic TAR RNA in oral epithelial cells. Cetuximab, a monoclonal neutralizing antibody against EGFR, blocks HIV-associated exosome-enhanced KSHV infection. Our findings reveal that saliva containing HIV-associated exosomes is a risk factor for the enhancement of KSHV infection and that the inhibition of EGFR serves as a novel strategy for preventing KSHV infection and transmission in the oral cavity.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is the causal agent for Kaposi's sarcoma (KS), the most common malignancy in HIV/AIDS patients. Oral transmission through saliva is considered the most common route for spreading the virus among HIV/AIDS patients. However, the role of HIV-specific components in the cotransfection of KSHV is unclear. We demonstrate that exosomes purified from the saliva of HIV-positive patients and secreted by HIV-infected T-cell lines promote KSHV infectivity in immortalized and primary oral epithelial cells. HIV-associated exosomes promote KSHV infection, which depends on HIV trans-activation response element (TAR) RNA and EGFR of oral epithelial cells, which can be targeted for reducing KSHV infection. These results reveal that HIV-associated exosomes are a risk factor for KSHV infection in the HIV-infected population.


Asunto(s)
Exosomas/metabolismo , Sarcoma de Kaposi/metabolismo , Adulto , Línea Celular , Epitelio/metabolismo , Epitelio/virología , Receptores ErbB/metabolismo , Infecciones por VIH/virología , VIH-1/metabolismo , VIH-1/fisiología , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/patogenicidad , Humanos , Masculino , Saliva/química , Saliva/virología , Sarcoma de Kaposi/virología , Activación Viral , Replicación Viral
2.
Cell Tissue Res ; 372(2): 427-431, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29470646

RESUMEN

Sleep apnea is a prevalent respiratory disease characterized by periodic cessation of breathing during sleep causing intermittent hypoxia (IH). Sleep apnea patients and rodents exposed to IH exhibit elevated sympathetic nerve activity and hypertension. A heightened carotid body (CB) chemoreflex has been implicated in causing autonomic abnormalities in IH-treated rodents and in sleep apnea patients. The purpose of this article is to review the emerging evidence showing that interactions between reactive oxygen species (ROS) and gaseous transmitters as a mechanism cause hyperactive CB by IH. Rodents treated with IH exhibit markedly elevated ROS in the CB, which is due to transcriptional upregulation of pro-oxidant enzymes by hypoxia-inducible factor (HIF)-1 and insufficient transcriptional regulation of anti-oxidant enzymes by HIF-2. ROS, in turn, increases cystathionine γ-lyase (CSE)-dependent H2S production in the CB. Blockade of H2S synthesis prevents IH-evoked CB activation. However, the effects of ROS on H2S production are not due to direct effects on CSE enzyme activity but rather due to inactivation of heme oxygenase-2 (HO-2), a carbon monoxide (CO) producing enzyme. CO inhibits H2S production through inactivation of CSE by PKG-dependent phosphorylation. During IH, reduced CO production resulting from inactivation of HO-2 by ROS releases the inhibition of CO on CSE thereby increasing H2S. Inhibiting H2S synthesis prevented IH-evoked sympathetic activation and hypertension.


Asunto(s)
Cuerpo Carotídeo/metabolismo , Cuerpo Carotídeo/patología , Gases/metabolismo , Hipoxia/metabolismo , Hipoxia/patología , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , Sulfuro de Hidrógeno/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(3): 1174-9, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24395806

RESUMEN

Oxygen (O2) sensing by the carotid body and its chemosensory reflex is critical for homeostatic regulation of breathing and blood pressure. Humans and animals exhibit substantial interindividual variation in this chemosensory reflex response, with profound effects on cardiorespiratory functions. However, the underlying mechanisms are not known. Here, we report that inherent variations in carotid body O2 sensing by carbon monoxide (CO)-sensitive hydrogen sulfide (H2S) signaling contribute to reflex variation in three genetically distinct rat strains. Compared with Sprague-Dawley (SD) rats, Brown-Norway (BN) rats exhibit impaired carotid body O2 sensing and develop pulmonary edema as a consequence of poor ventilatory adaptation to hypobaric hypoxia. Spontaneous Hypertensive (SH) rat carotid bodies display inherent hypersensitivity to hypoxia and develop hypertension. BN rat carotid bodies have naturally higher CO and lower H2S levels than SD rat, whereas SH carotid bodies have reduced CO and greater H2S generation. Higher CO levels in BN rats were associated with higher substrate affinity of the enzyme heme oxygenase 2, whereas SH rats present lower substrate affinity and, thus, reduced CO generation. Reducing CO levels in BN rat carotid bodies increased H2S generation, restoring O2 sensing and preventing hypoxia-induced pulmonary edema. Increasing CO levels in SH carotid bodies reduced H2S generation, preventing hypersensitivity to hypoxia and controlling hypertension in SH rats.


Asunto(s)
Monóxido de Carbono/química , Cuerpo Carotídeo/fisiología , Sulfuro de Hidrógeno/química , Hipertensión/metabolismo , Oxígeno/química , Edema Pulmonar/metabolismo , Animales , Peso Corporal , Catecolaminas/metabolismo , Cistationina gamma-Liasa/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Hipoxia , Inmunohistoquímica , Masculino , Consumo de Oxígeno , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Respiración , Transducción de Señal , Especificidad de la Especie , Nervios Esplácnicos/patología
4.
Proc Natl Acad Sci U S A ; 110(19): E1788-96, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610397

RESUMEN

Breathing and blood pressure are under constant homeostatic regulation to maintain optimal oxygen delivery to the tissues. Chemosensory reflexes initiated by the carotid body and catecholamine secretion from the adrenal medulla are the principal mechanisms for maintaining respiratory and cardiovascular homeostasis; however, the underlying molecular mechanisms are not known. Here, we report that balanced activity of hypoxia-inducible factor-1 (HIF-1) and HIF-2 is critical for oxygen sensing by the carotid body and adrenal medulla, and for their control of cardio-respiratory function. In Hif2α(+/-) mice, partial HIF-2α deficiency increased levels of HIF-1α and NADPH oxidase 2, leading to an oxidized intracellular redox state, exaggerated hypoxic sensitivity, and cardio-respiratory abnormalities, which were reversed by treatment with a HIF-1α inhibitor or a superoxide anion scavenger. Conversely, in Hif1α(+/-) mice, partial HIF-1α deficiency increased levels of HIF-2α and superoxide dismutase 2, leading to a reduced intracellular redox state, blunted oxygen sensing, and impaired carotid body and ventilatory responses to chronic hypoxia, which were corrected by treatment with a HIF-2α inhibitor. None of the abnormalities observed in Hif1α(+/-) mice or Hif2α(+/-) mice were observed in Hif1α(+/-);Hif2α(+/-) mice. These observations demonstrate that redox balance, which is determined by mutual antagonism between HIF-α isoforms, establishes the set point for hypoxic sensing by the carotid body and adrenal medulla, and is required for maintenance of cardio-respiratory homeostasis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cuerpo Carotídeo/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/metabolismo , Médula Suprarrenal/fisiología , Animales , Presión Sanguínea , Sistema Cardiovascular , Cuerpo Carotídeo/metabolismo , Catecolaminas/metabolismo , Heterocigoto , Homeostasis , Hipoxia , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Células PC12 , Ratas , Superóxido Dismutasa/metabolismo
5.
Am J Physiol Cell Physiol ; 308(2): C146-54, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25377087

RESUMEN

Arterial blood O2 levels are detected by specialized sensory organs called carotid bodies. Voltage-gated Ca(2+) channels (VGCCs) are important for carotid body O2 sensing. Given that T-type VGCCs contribute to nociceptive sensation, we hypothesized that they participate in carotid body O2 sensing. The rat carotid body expresses high levels of mRNA encoding the α1H-subunit, and α1H protein is localized to glomus cells, the primary O2-sensing cells in the chemoreceptor tissue, suggesting that CaV3.2 is the major T-type VGCC isoform expressed in the carotid body. Mibefradil and TTA-A2, selective blockers of the T-type VGCC, markedly attenuated elevation of hypoxia-evoked intracellular Ca(2+) concentration, secretion of catecholamines from glomus cells, and sensory excitation of the rat carotid body. Similar results were obtained in the carotid body and glomus cells from CaV3.2 knockout (Cacna1h(-/-)) mice. Since cystathionine-γ-lyase (CSE)-derived H2S is a critical mediator of the carotid body response to hypoxia, the role of T-type VGCCs in H2S-mediated O2 sensing was examined. Like hypoxia, NaHS, a H2S donor, increased intracellular Ca(2+) concentration and augmented carotid body sensory nerve activity in wild-type mice, and these effects were markedly attenuated in Cacna1h(-/-) mice. In wild-type mice, TTA-A2 markedly attenuated glomus cell and carotid body sensory nerve responses to hypoxia, and these effects were absent in CSE knockout mice. These results demonstrate that CaV3.2 T-type VGCCs contribute to the H2S-mediated carotid body response to hypoxia.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Canales de Calcio/metabolismo , Cuerpo Carotídeo/metabolismo , Sulfuro de Hidrógeno/farmacología , Hipoxia/metabolismo , Animales , Calcio/metabolismo , Cuerpo Carotídeo/efectos de los fármacos , Catecolaminas/metabolismo , Células Cultivadas , Células Quimiorreceptoras/efectos de los fármacos , Células Quimiorreceptoras/metabolismo , Cistationina gamma-Liasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/metabolismo , Ratas , Ratas Sprague-Dawley , Sulfuros
6.
Proc Natl Acad Sci U S A ; 109(7): 2515-20, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22232674

RESUMEN

Recurrent apnea with intermittent hypoxia is a major clinical problem in preterm infants. Recent studies, although limited, showed that adults who were born preterm exhibit increased incidence of sleep-disordered breathing and hypertension, suggesting that apnea of prematurity predisposes to autonomic dysfunction in adulthood. Here, we demonstrate that adult rats that were exposed to intermittent hypoxia as neonates exhibit exaggerated responses to hypoxia by the carotid body and adrenal chromaffin cells, which regulate cardio-respiratory function, resulting in irregular breathing with apneas and hypertension. The enhanced hypoxic sensitivity was associated with elevated oxidative stress, decreased expression of genes encoding antioxidant enzymes, and increased expression of pro-oxidant enzymes. Decreased expression of the Sod2 gene, which encodes the antioxidant enzyme superoxide dismutase 2, was associated with DNA hypermethylation of a single CpG dinucleotide close to the transcription start site. Treating neonatal rats with decitabine, an inhibitor of DNA methylation, during intermittent hypoxia exposure prevented oxidative stress, enhanced hypoxic sensitivity, and autonomic dysfunction. These findings implicate a hitherto uncharacterized role for DNA methylation in mediating neonatal programming of hypoxic sensitivity and the ensuing autonomic dysfunction in adulthood.


Asunto(s)
Epigénesis Genética , Corazón/fisiología , Homeostasis , Hipoxia/fisiopatología , Pulmón/fisiología , Animales , Animales Recién Nacidos , Islas de CpG , Metilación de ADN , Ratas
7.
Am J Physiol Cell Physiol ; 306(8): C745-52, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24477234

RESUMEN

The objective of the present study was to determine the impact of simulated apnea with intermittent hypoxia (IH) on endothelial barrier function and assess the underlying mechanism(s). Experiments were performed on human lung microvascular endothelial cells exposed to IH-consisting alternating cycles of 1.5% O2 for 30s followed by 20% O2 for 5 min. IH decreased transendothelial electrical resistance (TEER) suggesting attenuated endothelial barrier function. The effect of IH on TEER was stimulus dependent and reversible after reoxygenation. IH-exposed cells exhibited stress fiber formation and redistribution of cortactin, vascular endothelial-cadherins, and zona occludens-1 junction proteins along with increased intercellular gaps at cell-cell boundaries. Extracellular signal-regulated kinase (ERK) and c-jun NH2-terminal kinase (JNK) were phosphorylated in IH-exposed cells. Inhibiting either ERK or JNK prevented the IH-induced decrease in TEER and the reorganization of the cytoskeleton and junction proteins. IH increased reactive oxygen species (ROS) levels, and manganese (III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride, a membrane-permeable antioxidant, prevented ERK and JNK phosphorylation as well as IH-induced changes in endothelial barrier function. These results demonstrate that IH via ROS-dependent activation of MAP kinases leads to reorganization of cytoskeleton and junction proteins resulting in endothelial barrier dysfunction.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Oxígeno/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células Cultivadas , Citoesqueleto/fisiología , Impedancia Eléctrica , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Humanos , Hipoxia/metabolismo , Uniones Intercelulares/metabolismo , Pulmón/irrigación sanguínea , Estrés Oxidativo , Oxígeno/metabolismo , Fosforilación , Factores de Tiempo
8.
J Physiol ; 592(17): 3841-58, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24973414

RESUMEN

Previous studies reported that chronic intermittent hypoxia (CIH) results in an imbalanced expression of hypoxia-inducible factor-α (HIF-α) isoforms and oxidative stress in rodents, which may be due either to the direct effect of CIH or indirectly via hitherto uncharacterized mechanism(s). As neural activity is a potent regulator of gene transcription, we hypothesized that carotid body (CB) neural activity contributes to CIH-induced HIF-α isoform expression and oxidative stress in the chemoreflex pathway. Experiments were performed on adult rats exposed to CIH for 10 days. Rats exposed to CIH exhibited: increased HIF-1α and decreased HIF-2α expression; increased NADPH oxidase 2 and decreased superoxide dismutase 2 expression; and oxidative stress in the nucleus tractus solitarius and rostral ventrolateral medulla as well as in the adrenal medulla (AM), a major end organ of the sympathetic nervous system. Selective ablation of the CB abolished these effects. In the AM, sympathetic activation by the CB chemoreflex mediates CIH-induced HIF-α isoform imbalance via muscarinic acetylcholine receptor-mediated Ca(2+) influx, and the resultant activation of mammalian target of rapamycin pathway and calpain proteases. Rats exposed to CIH presented with hypertension, elevated sympathetic activity and increased circulating catecholamines. Selective ablation of either the CB (afferent pathway) or sympathetic innervation to the AM (efferent pathway) abolished these effects. These observations uncover CB neural activity-dependent regulation of HIF-α isoforms and the redox state by CIH in the central and peripheral nervous systems associated with the chemoreflex.


Asunto(s)
Cuerpo Carotídeo/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Estrés Oxidativo , Médula Suprarrenal/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Calcio/metabolismo , Calpaína/metabolismo , Hipoxia/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Reflejo , Núcleo Solitario/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
9.
Proc Natl Acad Sci U S A ; 108(7): 3065-70, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21288809

RESUMEN

Cardiorespiratory functions in mammals are exquisitely sensitive to changes in arterial O(2) levels. Hypoxia-inducible factors (e.g., HIF-1 and HIF-2) mediate transcriptional responses to reduced oxygen availability. We demonstrate that haploinsufficiency for the O(2)-regulated HIF-2α subunit results in augmented carotid body sensitivity to hypoxia, irregular breathing, apneas, hypertension, and elevated plasma norepinephrine levels in adult Hif-2α(+/-) mice. These dysregulated autonomic responses were associated with increased oxidative stress and decreased mitochondrial electron transport chain complex I activity in adrenal medullae as a result of decreased expression of major cytosolic and mitochondrial antioxidant enzymes. Systemic administration of a membrane-permeable antioxidant prevented oxidative stress, normalized hypoxic sensitivity of the carotid body, and restored autonomic functions in Hif-2α(+/-) mice. Thus, HIF-2α-dependent redox regulation is required for maintenance of carotid body function and cardiorespiratory homeostasis.


Asunto(s)
Médula Suprarrenal/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cuerpo Carotídeo/fisiología , Hipertensión/fisiopatología , Hipoxia/fisiopatología , Estrés Oxidativo/fisiología , Mecánica Respiratoria/fisiología , Análisis de Varianza , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Presión Sanguínea , Western Blotting , Complejo I de Transporte de Electrón/metabolismo , Perfilación de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Noqueados , Norepinefrina/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Pletismografía Total , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Toxins (Basel) ; 15(11)2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37999490

RESUMEN

The T-2 toxin (T2) poses a major threat to the health and productivity of animals. The present study aimed to investigate the regulatory mechanism of Nrf2 derived from broilers against T2-induced oxidative damage. DF-1 cells, including those with normal characteristics, as well as those overexpressing or with a knockout of specific components, were exposed to a 24 h treatment of 50 nM T2. The primary objective was to evaluate the indicators associated with oxidative stress and the expression of downstream antioxidant factors regulated by the Nrf2-ARE signaling pathway, at both the mRNA and protein levels. The findings of this study demonstrated a noteworthy relationship between the up-regulation of the Nrf2 protein and a considerable reduction in the oxidative stress levels within DF-1 cells (p < 0.05). Furthermore, this up-regulation was associated with a notable increase in the mRNA and protein levels of antioxidant factors downstream of the Nrf2-ARE signaling pathway (p < 0.05). Conversely, the down-regulation of the Nrf2 protein was linked to a marked elevation in oxidative stress levels in DF-1 cells (p < 0.05). Additionally, this down-regulation resulted in a significant decrease in both the mRNA and protein expression of antioxidant factors (p < 0.05). This experiment lays a theoretical foundation for investigating the detrimental impacts of T2 on broiler chickens. It also establishes a research framework for employing the Nrf2 protein in broiler chicken production and breeding. Moreover, it introduces novel insights for the prospective management of oxidative stress-related ailments in the livestock and poultry industry.


Asunto(s)
Antioxidantes , Toxina T-2 , Animales , Antioxidantes/farmacología , Antioxidantes/metabolismo , Pollos/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Toxina T-2/toxicidad , Toxina T-2/metabolismo , Estudios Prospectivos , Estrés Oxidativo , Transducción de Señal , Línea Celular , Fibroblastos/metabolismo , ARN Mensajero/metabolismo
11.
Toxins (Basel) ; 15(8)2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37624260

RESUMEN

In China, animal feeds are frequently contaminated with a range of mycotoxins, with Aflatoxin B1 (AFB1) and T-2 toxin (T-2) being two highly toxic mycotoxins. This study investigates the combined nephrotoxicity of AFB1 and T-2 on PK15 cells and murine renal tissues and their related oxidative stress mechanisms. PK15 cells were treated with the respective toxin concentrations for 24 h, and oxidative stress-related indicators were assessed. The results showed that the combination of AFB1 and T-2 led to more severe cellular damage and oxidative stress compared to exposure to the individual toxins (p < 0.05). In the in vivo study, pathological examination revealed that the kidney tissue of mice exposed to the combined toxins showed signs of glomerular atrophy. The contents of oxidative stress-related indicators were significantly increased in the kidney tissue (p < 0.05). These findings suggest that the combined toxins cause significant oxidative damage to mouse kidneys. The study highlights the importance of considering the combined effects of mycotoxins in animal feed, particularly AFB1 and T-2, which can lead to severe nephrotoxicity and oxidative stress in PK15 cells and mouse kidneys. The findings have important implications for animal feed safety and regulatory policy.


Asunto(s)
Micotoxinas , Toxinas Biológicas , Animales , Ratones , Aflatoxina B1/toxicidad , Glomérulos Renales , Estrés Oxidativo
12.
Proc Natl Acad Sci U S A ; 106(4): 1199-204, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19147445

RESUMEN

Intermittent hypoxia (IH) occurs in many pathological conditions including recurrent apneas. Hypoxia-inducible factors (HIFs) 1 and 2 mediate transcriptional responses to low O(2). A previous study showed that HIF-1 mediates some of the IH-evoked physiological responses. Because HIF-2alpha is an orthologue of HIF-1alpha, we examined the effects of IH on HIF-2alpha, the O(2)-regulated subunit expression, in pheochromocytoma 12 cell cultures. In contrast to the up-regulation of HIF-1alpha, HIF-2alpha was down-regulated by IH. Similar down-regulation of HIF-2alpha was also seen in carotid bodies and adrenal medullae from IH-exposed rats. Inhibitors of calpain proteases (ALLM, ALLN) prevented IH-evoked degradation of HIF-2alpha whereas inhibitors of prolyl hydroxylases or proteosome were ineffective. IH activated calpain proteases and down-regulated the endogenous calpain inhibitor calpastatin. IH-evoked HIF-2alpha degradation led to inhibition of SOD2 transcription, resulting in oxidative stress. Over-expression of transcriptionally active HIF-2alpha prevented IH-evoked oxidative stress and restored SOD2 activity. Systemic treatment of IH-exposed rats with ALLM rescued HIF-2alpha degradation and restored SOD2 activity, thereby preventing oxidative stress and hypertension. These observations demonstrate that, unlike continuous hypoxia, IH leads to down-regulation of HIF-2alpha via a calpain-dependent signaling pathway and results in oxidative stress as well as autonomic morbidities.


Asunto(s)
Apnea/enzimología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Calpaína/metabolismo , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Animales , Apnea/mortalidad , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/patología , Señalización del Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Calpaína/antagonistas & inhibidores , Hipoxia de la Célula/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/antagonistas & inhibidores
13.
Sci Rep ; 12(1): 21779, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36526691

RESUMEN

Elevated serum cytokine production in COVID-19 patients is associated with disease progression and severity. However, the stimuli that initiate cytokine production in patients remain to be fully revealed. Virus-infected cells release virus-associated exosomes, extracellular vesicles of endocytic origin, into the blood to deliver viral cargoes able to regulate immune responses. Here, we report that plasma exosomes of COVID-19 patients contain SARS-CoV-2 double stranded RNA (dsRNA) and stimulate robust production of interleukin-6 (IL-6), IL-8, tumor necrosis factor-α (TNF-α), and other inflammatory cytokines and chemokines by human peripheral mononuclear cells. Exosome depletion abolished these stimulated responses. COVID-19 plasma exosomes induced proinflammatory responses in CD4+ T cells, CD8+ T cells, and CD14+ monocytes but not significantly in regulatory T cells, Th17 T cells, or central memory T cells. COVID-19 plasma exosomes protect the SARS-CoV-2 dsRNA cargo from RNase and deliver the dsRNA into recipient cells. These exosomes significantly increase expression of endosomal toll-like receptor 3 (TLR3), TLR7, TLR8, and TLR9 in peripheral T cells and monocytes. A pharmacological inhibitor of TLR3 considerably reduced cytokine and chemokine production by CD4+ and CD8+ T cells but not by CD14+ monocytes, highlighting divergent signaling pathways of immune cells in response to COVID-19 plasma exosomes. Our results identify a novel model of intercellular crosstalk following SARS-CoV-2 infection that evoke immune responses positioned to contribute to elevated cytokine production associated with COVID-19 progression, severity, and long-haul symptoms.


Asunto(s)
COVID-19 , Exosomas , Humanos , Exosomas/metabolismo , Receptor Toll-Like 3/metabolismo , Leucocitos Mononucleares/metabolismo , Linfocitos T CD8-positivos/metabolismo , SARS-CoV-2/metabolismo , COVID-19/metabolismo , Citocinas/metabolismo , ARN Bicatenario/metabolismo , Inmunidad
14.
J Neurosci ; 30(32): 10763-72, 2010 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-20705601

RESUMEN

Nearly 90% of premature infants experience the stress of intermittent hypoxia (IH) as a consequence of recurrent apneas (periodic cessation of breathing). In neonates, catecholamine secretion from the adrenal medulla is critical for maintaining homeostasis under hypoxic stress. We recently reported that IH treatment enhanced hypoxia-evoked catecholamine secretion and [Ca2+]i responses in neonatal rat adrenal chromaffin cells and involves reactive oxygen species (ROS). The purpose of the present study was to identify the source(s) of ROS generation and examine the mechanisms underlying the enhanced catecholamine secretion by IH. Neonatal rats of either sex (postal day 0-5) were exposed to either IH or normoxia. IH treatment increased NADPH oxidase (NOX) activity, upregulated NOX2 and NOX4 transcription in adrenal medullae, and a NOX inhibitor prevented the effects of IH on hypoxia-evoked chromaffin cell secretion. IH upregulated Cav3.1 and Cav3.2 T-type Ca2+ channel mRNAs via NOX/ROS signaling and augmented T-type Ca2+ current in IH-treated chromaffin cells. Mibefradil, a blocker of T-type Ca2+ channels attenuated the effects of hypoxia on [Ca2+]i and catecholamine secretion in IH-treated cells. In Ca2+-free medium, IH-treated cells exhibited higher basal [Ca2+]i levels and more pronounced [Ca2+]i responses to hypoxia compared with controls, and blockade of ryanodine receptors (RyRs) prevented these effects. RyR2 and RyR3 mRNAs were upregulated, RyR2 was S-glutathionylated in IH-treated adrenal medullae, and NOX/ROS inhibitors prevented these effects. These results demonstrate that neonatal IH treatment leads to NOX/ROS-dependent recruitment of T-type Ca2+ channels and RyRs, resulting in augmented [Ca2+]i mobilization and catecholamine secretion.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Catecolaminas/metabolismo , Células Cromafines/fisiología , Exocitosis/fisiología , Hipoxia/patología , NADPH Oxidasas/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Animales Recién Nacidos , Cloruro de Cadmio/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/genética , Células Cromafines/efectos de los fármacos , Células Cromafines/metabolismo , Electroquímica/métodos , Inhibidores Enzimáticos/farmacología , Exocitosis/efectos de los fármacos , Femenino , Hipoxia/fisiopatología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Ratas , Canal Liberador de Calcio Receptor de Rianodina/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
15.
J Cell Physiol ; 226(11): 2925-33, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21302291

RESUMEN

Sleep-disordered breathing with recurrent apnea is associated with intermittent hypoxia (IH). Cardiovascular morbidities caused by IH are triggered by increased generation of reactive oxygen species (ROS) by pro-oxidant enzymes, especially NADPH oxidase-2 (Nox2). Previous studies showed that (i) IH activates hypoxia-inducible factor 1 (HIF-1) in a ROS-dependent manner and (ii) HIF-1 is required for IH-induced ROS generation, indicating the existence of a feed-forward mechanism. In the present study, using multiple pharmacological and genetic approaches, we investigated whether IH-induced expression of Nox2 is mediated by HIF-1 in the central and peripheral nervous system of mice as well as in cultured cells. IH increased Nox2 mRNA, protein, and enzyme activity in PC12 pheochromocytoma cells as well as in wild-type mouse embryonic fibroblasts (MEFs). This effect was abolished or attenuated by blocking HIF-1 activity through RNA interference or pharmacologic inhibition (digoxin or YC-1) or by genetic knockout of HIF-1α in MEFs. Increasing HIF-1α expression by treating PC 12 cells with the iron chelator deferoxamine for 20 h or by transfecting them with HIF-1alpha expression vector increased Nox2 expression and enzyme activity. Exposure of wild-type mice to IH (8 h/day for 10 days) up-regulated Nox2 mRNA expression in brain cortex, brain stem, and carotid body but not in cerebellum. IH did not induce Nox2 expression in cortex, brainstem, carotid body, or cerebellum of Hif1a(+/-) mice, which do not manifest increased ROS or cardiovascular morbidities in response to IH. These results establish a pathogenic mechanism linking HIF-1, ROS generation, and cardiovascular pathology in response to IH.


Asunto(s)
Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Glicoproteínas de Membrana/genética , NADPH Oxidasas/genética , Especies Reactivas de Oxígeno/metabolismo , Síndromes de la Apnea del Sueño/metabolismo , Animales , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/enzimología , Deferoxamina/farmacología , Digoxina/farmacología , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Furanos/farmacología , Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Indazoles/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , NADPH Oxidasa 2 , Células PC12 , Sistema Nervioso Periférico/efectos de los fármacos , Sistema Nervioso Periférico/enzimología , Ratas , Sideróforos/farmacología , Síndromes de la Apnea del Sueño/genética
16.
Am J Physiol Cell Physiol ; 299(2): C381-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20664070

RESUMEN

We recently reported that adrenomedullary chromaffin cells (AMC) from neonatal rats treated with intermittent hypoxia (IH) exhibit enhanced catecholamine secretion by hypoxia (Souvannakitti D, Kumar GK, Fox A, Prabhakar NR. J Neurophysiol 101: 2837-2846, 2009). In the present study, we examined whether neonatal IH also facilitate AMC responses to nicotine, a potent stimulus to chromaffin cells. Experiments were performed on rats exposed to either IH (15-s hypoxia-5-min normoxia; 8 h/day) or to room air (normoxia; controls) from ages postnatal day 0 (P0) to P5. Quantitative RT-PCR analysis revealed expression of mRNAs alpha(3-), alpha(5-), alpha(7-), and beta(2-) and beta(4-)nicotinic acetylcholine receptor (nAChR) subunits in adrenal medullae from control P5 rats. Nicotine-elevated intracellular Ca(2+) concentration ([Ca(2+)](i)) in AMC and nAChR antagonists prevented this response, suggesting that nAChRs are functional in neonatal AMC. In IH-treated rats, nAChR mRNAs were downregulated in AMC, which resulted in a markedly attenuated nicotine-evoked elevation in [Ca(2+)](i) and subsequent catecholamine secretion. Systemic administration of antioxidant prevented IH-evoked downregulation of nAChR expression and function. P35 rats treated with neonatal IH exhibited reduced nAChR mRNA expression in adrenal medullae, attenuated AMC responses to nicotine, and impaired neurogenic catecholamine secretion. Thus the response to neonatal IH lasts for at least 30 days. These observations demonstrate that neonatal IH downregulates nAChR expression and function in AMC via reactive oxygen species signaling, and the effects of neonatal IH persist at least into juvenile life, leading to impaired neurogenic catecholamine secretion from AMC.


Asunto(s)
Células Cromafines/metabolismo , Hipoxia/metabolismo , Receptores Nicotínicos/biosíntesis , Animales , Animales Recién Nacidos , Antioxidantes/farmacología , Calcio/metabolismo , Células Cultivadas , Células Cromafines/fisiología , Femenino , Regulación de la Expresión Génica , Nicotina/farmacología , Oxígeno/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/genética , Receptores Nicotínicos/fisiología
17.
J Cell Physiol ; 217(3): 674-85, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18651560

RESUMEN

Sleep-disordered breathing with recurrent apnea (periodic cessation of breathing) results in chronic intermittent hypoxia (IH), which leads to cardiovascular and respiratory pathology. Molecular mechanisms underlying IH-evoked cardio-respiratory co-morbidities have not been delineated. Mice with heterozygous deficiency of hypoxia-inducible factor 1alpha (HIF-1alpha) do not develop cardio-respiratory responses to chronic IH. HIF-1alpha protein expression and HIF-1 transcriptional activity are induced by IH in PC12 cells. In the present study, we investigated the signaling pathways associated with IH-evoked HIF-1alpha accumulation. PC12 cells were exposed to aerobic conditions (20% O(2)) or 60 cycles of IH (30 sec at 1.5% O(2) followed by 5 min at 20% O(2)). Our results show that IH-induced HIF-1alpha accumulation is due to increased generation of ROS by NADPH oxidase. We further demonstrate that ROS-dependent Ca(2+) signaling pathways involving phospholipase Cgamma (PLCgamma) and protein kinase C activation are required for IH-evoked HIF-1alpha accumulation. IH leads to activation of mTOR and S6 kinase (S6K) and rapamycin partially inhibited IH-induced HIF-1alpha accumulation. IH also decreased hydroxylation of HIF-1alpha protein and anti-oxidants as well as inhibitors of Ca(+2) signaling prevented this response. Thus, both increased mTOR-dependent HIF-1alpha synthesis and decreased hydroxylase-dependent HIF-1alpha degradation contribute to IH-evoked HIF-1alpha accumulation. Following IH, HIF-1alpha, and phosphorylated mTOR levels remained elevated during 90 min of re-oxygenation despite re-activation of prolyl hydroxylase. Rapamycin or cycloheximide, blocked increased HIF-1alpha levels during re-oxygenation indicating that mTOR-dependent protein synthesis is required for the persistent elevation of HIF-1alpha levels during re-oxygenation.


Asunto(s)
Señalización del Calcio , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , NADPH Oxidasas/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteínas Quinasas/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Hidroxilación/efectos de los fármacos , Ratones , Modelos Biológicos , Oxígeno/farmacología , Células PC12 , Fosfolipasa C gamma/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Termodinámica
18.
Biochem Biophys Res Commun ; 373(2): 309-14, 2008 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-18570888

RESUMEN

Previous studies suggest that reactive oxygen species (ROS) play an important role in physiological responses to hypoxia. In the present study, we examined the effects of hypoxia on human ether-a-go-go related gene (hERG) channel protein expression and assessed the role of ROS. Hypoxia, in a stimulus- and time-dependent manner, decreased hERG protein with marked reduction in hERG K+ conductance in human embryonic kidney cells stably expressing the hERG alpha subunit. Down-regulation of hERG by hypoxia was not due to increased proteasomal degradation or decreased transcription but due to decreased synthesis of the protein. Hypoxia increased ROS in a time-dependent manner. Antioxidants prevented hypoxia-evoked down-regulation of hERG protein and exogenous oxidants mimicked the effects of hypoxia. Hypoxia-evoked down-regulation of hERG protein and elevation in ROS were absent in p(O) cells, which are devoid of mitochondrial DNA. Inhibitors of NADPH oxidase failed to prevent the effects of hypoxia. These results demonstrate that hypoxia enhances the production of ROS in the mitochondria, resulting in down-regulation of hERG translation and decreased hERG-mediated K+ conductance.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Mitocondrias/metabolismo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Anaerobiosis , Antioxidantes/farmacología , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/biosíntesis , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Biosíntesis de Proteínas
19.
Respir Physiol Neurobiol ; 164(1-2): 277-81, 2008 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-18692603

RESUMEN

Recurrent apneas are characterized by transient repetitive cessations of breathing (two breaths duration or longer) resulting in periodic decreases in arterial blood PO2 or chronic intermittent hypoxia (IH). Patients with recurrent apneas and experimental animals exposed to chronic IH exhibit cardio-respiratory morbidities. The purpose of this article is to highlight the current information on the transcriptional mechanisms associated with chronic IH. Studies on rodents and cell cultures have shown that IH activates a variety of transcription factors including the hypoxia-inducible factor-1 (HIF-1), c-fos (immediate early gene), nuclear factor of activated T-cells (NFAT), and nuclear factor kB (NF-kB). The signaling pathways associated with transcriptional activation associated with IH differ from continuous hypoxia (CH). Compared to same duration and intensity of CH, IH is more potent in activating HIF-1 and c-fos and also results in long-lasting accumulation of HIF-1alpha and c-fos mRNA, a phenomenon that was not seen with CH. IH-evoked transcriptional activation by HIF-1, c-fos as well as the resulting activator protein-1 (AP-1) requires reactive oxygen species (ROS)-mediated signaling and involves complex feed forward interactions between HIF-1 and ROS. Chronic IH-evoked cardio-respiratory responses are absent in Hif-1alpha+/- mice, and hypertension elicited by chronic IH is absent in mice lacking NFAT3c. These studies indicate that cardiorespiratory responses to chronic IH depend on complex interactions between various transcription factors resulting in alterations in several down stream genes and their protein products.


Asunto(s)
Hipoxia/fisiopatología , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología , Animales , Humanos , Hipoxia/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/genética
20.
Guang Pu Xue Yu Guang Pu Fen Xi ; 27(2): 287-91, 2007 Feb.
Artículo en Zh | MEDLINE | ID: mdl-17514957

RESUMEN

A batch of potassium lithium niobate (KLN) crystals with different compositions were grown by using TSSG technique. Samples with three different compositions were well polished. By using near infrared cw:Ti-sapphire laser, their Second Harmonic Generation (SHG) properties were investigated. The results showed that the SHG effect is related to the composition of the samples, and their frequency-doubling efficiency enhanced with the raise of Li ions content in the crystal. By using infrared Raman technique, the properties of nonlinear lattice vibration of thee samples were investigated, and the character of Raman spectrum were analyzed, as well the effect of composition on the SHG properties were discussed. The analysis results showed the striking effects of Li content for these Raman peaks. For KLN sample with small Li content, the three character peaks belonged to [NbO6]7- octahedron show simple peak. With the raise of Li content in crystal, the peaks belonged to v2 mode were partly split, and the peak belongs to v5 mode was broadened. When the Li content approach to the chemical composition KLN crystal, and the structure of [NbO6]7- octahedron is almost to be disorganized, the peaks belonged to v5 mode were split, and the peaks belonged to v1 mode and v2 mode were partly split also, with more distinct weak peaks in the wavelength range of 100-400 cm(-1). These effects were caused by the raise of Li content, which leads to the severer aberrance of [NbO6]7- octahedron in KLN crystal, and disturbing the lattice vibration of the octahedron. This phenomenon is agreed with the nonlinear properties of potassium lithium niobate crystal.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA