Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 15360, 2023 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-37717072

RESUMEN

Sleep restriction is associated with increased cardiovascular risk, which is more pronounced in female than male persons. We reported recently first causal evidence that mild, prolonged sleep restriction mimicking "real-life" conditions impairs endothelial function, a key step in the development and progression of cardiovascular disease, in healthy female persons. However, the underlying mechanisms are unclear. In model organisms, sleep restriction increases oxidative stress and upregulates antioxidant response via induction of the antioxidant regulator nuclear factor (erythroid-derived 2)-like 2 (Nrf2). Here, we assessed directly endothelial cell oxidative stress and antioxidant responses in healthy female persons (n = 35) after 6 weeks of mild sleep restriction (1.5 h less than habitual sleep) using randomized crossover design. Sleep restriction markedly increased endothelial oxidative stress without upregulating antioxidant response. Using RNA-seq and a predicted protein-protein interaction database, we identified reduced expression of endothelial Defective in Cullin Neddylation-1 Domain Containing 3 (DCUN1D3), a protein that licenses Nrf2 antioxidant responses, as a mediator of impaired endothelial antioxidant response in sleep restriction. Thus, sleep restriction impairs clearance of endothelial oxidative stress that over time increases cardiovascular risk.Trial Registration: NCT02835261 .


Asunto(s)
Antioxidantes , Enfermedades Cardiovasculares , Humanos , Femenino , Masculino , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Células Endoteliales , Enfermedades Cardiovasculares/etiología
2.
Am J Physiol Lung Cell Mol Physiol ; 325(3): L277-L287, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37431588

RESUMEN

Failure of the lung's endothelial barrier underlies lung injury, which causes the high mortality acute respiratory distress syndrome (ARDS). Multiple organ failure predisposes to the mortality, but mechanisms are poorly understood. Here, we show that mitochondrial uncoupling protein 2 (UCP2), a component of the mitochondrial inner membrane, plays a role in the barrier failure. Subsequent lung-liver cross talk mediated by neutrophil activation causes liver congestion. We intranasally instilled lipopolysaccharide (LPS). Then, we viewed the lung endothelium by real-time confocal imaging of the isolated, blood-perfused mouse lung. LPS caused alveolar-capillary transfer of reactive oxygen species and mitochondrial depolarization in lung venular capillaries. The mitochondrial depolarization was inhibited by transfection of alveolar Catalase and vascular knockdown of UCP2. LPS instillation caused lung injury as indicated by increases in bronchoalveolar lavage (BAL) protein content and extravascular lung water. LPS or Pseudomonas aeruginosa instillation also caused liver congestion, quantified by liver hemoglobin and plasma aspartate aminotransferase (AST) increases. Genetic inhibition of vascular UCP2 prevented both lung injury and liver congestion. Antibody-mediated neutrophil depletion blocked the liver responses, but not lung injury. Knockdown of lung vascular UCP2 mitigated P. aeruginosa-induced mortality. Together, these data suggest a mechanism in which bacterial pneumonia induces oxidative signaling to lung venular capillaries, known sites of inflammatory signaling in the lung microvasculature, depolarizing venular mitochondria. Successive activation of neutrophils induces liver congestion. We conclude that oxidant-induced UCP2 expression in lung venular capillaries causes a mechanistic sequence leading to liver congestion and mortality. Lung vascular UCP2 may present a therapeutic target in ARDS.NEW & NOTEWORTHY We report that mitochondrial injury in lung venular capillaries underlies barrier failure in pneumonia, and venular capillary uncoupling protein 2 (UCP2) causes neutrophil-mediated liver congestion. Using in situ imaging, we found that epithelial-endothelial transfer of H2O2 activates UCP2, depolarizing mitochondria in venular capillaries. The conceptual advance from our findings is that mitochondrial depolarization in lung capillaries mediates liver cross talk through circulating neutrophils. Pharmacologic blockade of UCP2 could be a therapeutic strategy for lung injury.


Asunto(s)
Lesión Pulmonar , Neumonía Bacteriana , Síndrome de Dificultad Respiratoria , Ratones , Animales , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Capilares/metabolismo , Peróxido de Hidrógeno , Hígado/metabolismo , Mitocondrias/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Lesión Pulmonar/metabolismo , Neumonía Bacteriana/metabolismo , Proteínas Mitocondriales/metabolismo
3.
Ann Am Thorac Soc ; 20(7): 1029-1037, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36912897

RESUMEN

Rationale: Increased cardiovascular risk in obstructive sleep apnea (OSA) persists after continuous positive airway pressure (CPAP) and alternative therapies are needed. Impaired endothelial protection against complement is a cholesterol-dependent process that initiates endothelial inflammation in OSA, which increases cardiovascular risk. Objectives: To investigate directly whether lowering cholesterol improves endothelial protection against complement and its proinflammatory effects in OSA. Methods: Newly diagnosed patients with OSA (n = 87) and OSA-free controls (n = 32) participated. Endothelial cells and blood were collected at baseline, after 4 weeks of CPAP therapy, and again after 4 weeks of 10 mg atorvastatin versus placebo using a randomized, double-blind, parallel-group design. Primary outcome was the proportion of a complement inhibitor, CD59, on the endothelial cell plasma membrane in OSA patients after 4 weeks of statins versus placebo. Secondary outcomes were complement deposition on endothelial cells and circulating levels of its downstream proinflammatory factor, angiopoietin-2, after statins versus placebo. Results: Baseline expression of CD59 was lower, whereas complement deposition on endothelial cells and levels of angiopoietin-2 were greater, in patients with OSA compared with controls. CPAP did not affect expression of CD59 or complement deposition on endothelial cells in patients with OSA, regardless of adherence. Compared with placebo, statins increased expression of endothelial complement protector CD59 and lowered complement deposition in patients with OSA. Good CPAP adherence was associated with increased angiopoietin-2 levels, which was reversed by statins. Conclusions: Statins restore endothelial protection against complement and reduce its downstream proinflammatory effects, suggesting a potential approach to reduce residual cardiovascular risk after CPAP in patients with OSA. Clinical trial registered with www.clinicaltrials.gov (NCT03122639).


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Apnea Obstructiva del Sueño , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Angiopoyetina 2 , Células Endoteliales , Colesterol , Presión de las Vías Aéreas Positiva Contínua , Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/terapia
5.
Sleep ; 44(4)2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33351148

RESUMEN

STUDY OBJECTIVE: Obstructive sleep apnea (OSA) is highly prevalent and triples vascular thromboembolic risk. Intermittent hypoxia (IH) during transient cessation of breathing in OSA impairs endothelial protection against complement. Complement activation stimulates the endothelial release of a pro-thrombotic von Willebrand factor (vWF). We investigated whether increased complement activity in OSA promotes the endothelial release of vWF and pro-inflammatory angiopoietin-2. We further investigated whether improving complement protection with statins reverses these changes. METHODS: Using endothelial cells (ECs) and blood collected from OSA patients (n = 109) and controls (n = 67), we assessed whether altered cellular localization of complement inhibitor CD59 in OSA modulates exocytosis of Weibel-Palade bodies (WPB), secretory granules that store vWF and angiopoietin-2. These interactions were also assessed in vitro in ECs exposed to normoxia or IH with or without recombinant complement C9 and with or without atorvastatin. RESULTS: Circulating levels of angiopoietin-2 were greater in OSA than controls and levels of vWF cleavage products correlated with OSA severity. In cultured ECs, IH enhanced complement-stimulated angiopoietin-2 and vWF release by reducing EC surface and increasing intracellular expression of complement inhibitor CD59. Intracellular CD59 co-localized with WPB in OSA. IH increased binding of intracellular CD59 to syntaxin-3, which dissociated syntaxin-3 from voltage-sensitive calcium channel Cav1.2, and activated WPB exocytosis in a calcium-dependent manner. Atorvastatin reversed IH-enhanced endothelial release of vWF and angiopoietin-2. CONCLUSIONS: IH promotes the complement-mediated release of vWF and angiopoietin-2, which may contribute to pro-thrombotic and pro-inflammatory conditions in OSA. Statin reversed these effects, suggesting a potential approach to reduce cardiovascular risk in OSA.


Asunto(s)
Apnea Obstructiva del Sueño , Factor de von Willebrand , Angiopoyetina 2 , Células Cultivadas , Células Endoteliales , Humanos , Cuerpos de Weibel-Palade
6.
Nutr Res ; 75: 77-84, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32045786

RESUMEN

The Mediterranean Diet, characterized by higher intakes of plant foods including plant proteins, monounsaturated fat, fish, and lower consumption of animal products and saturated fat, has long been associated with reduced cardiovascular risk, but the molecular mechanisms underlying these associations have not been fully elucidated. We conducted a pilot study to evaluate associations of an Alternate Mediterranean Diet Score, reflective of adherence to this diet pattern and adapted for US populations, and its components, with markers of endothelial inflammation directly measured in endothelial cells harvested from a diverse sample of women (n = 25, mean ±â€¯SD age 33 ±â€¯10.5y, 68% racial/ethnic minorities). Cardiovascular risk markers including nuclear factor kappa B (NF-κB)-a marker of inflammation, as well as oxidative stress and endothelial nitric oxide synthase (eNOS) gene expression-markers of endothelial function, were evaluated in harvested endothelial cells. We hypothesized that the Mediterranean diet pattern would be associated with lower inflammation and oxidative stress and higher eNOS expression in endothelial cells. Results showed that lower oxidative stress was associated with higher plant-based protein (Exp(ß) = 0.96; P = .007), overall protein (Exp(ß) = 0.99; P = .007), and red and processed meat intake (Exp(ß) = 0.93; P = .012). Lower NF-κB was associated with higher legume (Exp(ß) = 0.79; P = .045) intake, and higher eNOS was associated with higher red and processed meat intake (Exp(ß) = 1.13; P = .005). Our findings suggest potential novel mechanisms through which certain Mediterranean dietary components may influence pre-clinical vascular alterations that may be associated with cardiovascular risk through lower endothelial oxidative stress, lower inflammation, and greater endothelial functioning. These findings warrant confirmation, prospectively in a larger sample.


Asunto(s)
Dieta Mediterránea , Células Endoteliales/fisiología , Etnicidad , Inflamación/prevención & control , Proyectos Piloto , Adulto , Dieta Vegetariana , Células Endoteliales/química , Endotelio Vascular/fisiopatología , Fabaceae , Femenino , Expresión Génica , Humanos , Inflamación/fisiopatología , FN-kappa B/análisis , Óxido Nítrico Sintasa de Tipo III/genética , Estrés Oxidativo/fisiología , Proteínas de Plantas/administración & dosificación , Estados Unidos
7.
J Am Heart Assoc ; 7(12)2018 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-29886425

RESUMEN

BACKGROUND: Insufficient sleep increases blood pressure. However, the effects of milder, highly prevalent but frequently neglected sleep disturbances, including poor sleep quality and insomnia, on vascular health in women are unclear. We investigated whether poor sleep patterns are associated with blood pressure and endothelial inflammation in a diverse sample of women. METHODS AND RESULTS: Women who participated in the ongoing American Heart Association Go Red for Women Strategically Focused Research Network were studied (n=323, 57% minority, mean age=39±17 years, range=20-79 years). Sleep duration, sleep quality, and time to sleep onset were assessed using the Pittsburgh Sleep Quality Index (score ≥5=poor sleep quality). Risk for obstructive sleep apnea was evaluated using the Berlin questionnaire, and insomnia was assessed using the Insomnia Severity Index. In a subset of women who participated in the basic study (n=26), sleep duration was assessed objectively using actigraphy, and endothelial inflammation was assessed directly in harvested endothelial cells by measuring nuclear translocation of nuclear factor kappa B. Vascular reactivity was measured by brachial artery flow-mediated dilation (n=26). Systolic and diastolic blood pressure were measured by trained personnel (n=323). Multivariable linear regressions were used to evaluate associations between sleep patterns and blood pressure, nuclear factor kappa B, and flow-mediated dilation. Mean sleep duration was 6.8±1.3 hours/night in the population study and 7.5±1.1 hour/night in the basic study. In the population study sample, 50% had poor sleep quality versus 23% in the basic study, and 37% had some level of insomnia versus 15% in the basic study. Systolic blood pressure was associated directly with poor sleep quality, and diastolic blood pressure was of borderline significance with obstructive sleep apnea risk after adjusting for confounders (P=0.04 and P=0.08, respectively). Poor sleep quality was associated with endothelial nuclear factor kappa B activation (ß=30.6; P=0.03). Insomnia and longer sleep onset latency were also associated with endothelial nuclear factor kappa B activation (ß=27.6; P=0.002 and ß=8.26; P=0.02, respectively). No evidence was found for an association between sleep and flow-mediated dilation. CONCLUSIONS: These findings provide direct evidence that common but frequently neglected sleep disturbances such as poor sleep quality and insomnia are associated with increased blood pressure and vascular inflammation even in the absence of inadequate sleep duration in women. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02835261.


Asunto(s)
Presión Sanguínea , Endotelio Vascular/fisiopatología , Hipertensión/epidemiología , Inflamación/epidemiología , Trastornos del Inicio y del Mantenimiento del Sueño/epidemiología , Sueño , Salud de la Mujer , Adulto , Anciano , American Heart Association , Biomarcadores/sangre , Endotelio Vascular/metabolismo , Femenino , Humanos , Hipertensión/fisiopatología , Inflamación/sangre , Inflamación/fisiopatología , Persona de Mediana Edad , FN-kappa B/sangre , Prevalencia , Pronóstico , Factores de Riesgo , Factores Sexuales , Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología , Factores de Tiempo , Estados Unidos/epidemiología , Vasodilatación , Adulto Joven
8.
Sci Transl Med ; 8(320): 320ra1, 2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26738794

RESUMEN

Obstructive sleep apnea (OSA), characterized by intermittent hypoxia (IH) during transient cessation of breathing, triples the risk for cardiovascular diseases. We used a phage display peptide library as an unbiased approach to investigate whether IH, which is specific to OSA, activates endothelial cells (ECs) in a distinctive manner. The target of a differentially bound peptide on ECs collected from OSA patients was identified as CD59, a major complement inhibitor that protects ECs from the membrane attack complex (MAC). A decreased proportion of CD59 is located on the EC surface in OSA patients compared with controls, suggesting reduced protection against complement attack. In vitro, IH promoted endothelial inflammation predominantly via augmented internalization of CD59 and consequent MAC deposition. Increased internalization of endothelial CD59 in IH appeared to be cholesterol-dependent and was reversed by statins in a CD59-dependent manner. These studies suggest that reduced complement inhibition may mediate endothelial inflammation and increase vascular risk in OSA patients.


Asunto(s)
Antígenos CD59/metabolismo , Endocitosis , Endotelio Vascular/patología , Inflamación/patología , Apnea Obstructiva del Sueño/patología , Transporte Biológico/efectos de los fármacos , Estudios de Casos y Controles , Técnicas de Visualización de Superficie Celular , Colesterol/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Endocitosis/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipoxia/complicaciones , Hipoxia/patología , Proteínas de la Membrana/metabolismo , Apnea Obstructiva del Sueño/complicaciones
9.
Chest ; 148(1): 112-119, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25742439

RESUMEN

BACKGROUND: The relative risk for cardiovascular diseases in passive smokers is similar to that of active smokers despite almost a 100-fold lower dose of inhaled cigarette smoke. However, the mechanisms underlying the surprising susceptibility of the vascular tissue to the toxins in secondhand smoke (SHS) have not been directly investigated. The aim of this study was to investigate directly vascular endothelial cell function in passive smokers. METHODS: Using a minimally invasive method of endothelial biopsy, we investigated directly the vascular endothelium in 23 healthy passive smokers, 25 healthy active smokers, and 23 healthy control subjects who had never smoked and had no regular exposure to SHS. Endothelial nitric oxide synthase (eNOS) function (expression of basal eNOS and activated eNOS [phosphorylated eNOS at serine1177 (P-eNOS)]) and expression of markers of inflammation (nuclear factor-κB [NF-κB]) and oxidative stress (nitrotyrosine) were assessed in freshly harvested venous endothelial cells by quantitative immunofluorescence. RESULTS: Expression of eNOS and P-eNOS was similarly reduced and expression of NF-κB was similarly increased in passive and active smokers compared with control subjects. Expression of nitrotyrosine was greater in active smokers than control subjects and similar in passive and active smokers. Brachial artery flow-mediated dilation was similarly reduced in passive and active smokers compared with control subjects, consistent with reduced endothelial NO bioavailability. CONCLUSIONS: Secondhand smoking increases vascular endothelial inflammation and reduces active eNOS to a similar extent as active cigarette smoking, indicating direct toxic effects of SHS on the vasculature.


Asunto(s)
Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Fumar/patología , Contaminación por Humo de Tabaco/efectos adversos , Adulto , Arteria Braquial/fisiología , Estudios de Casos y Controles , Cotinina/sangre , Células Endoteliales/patología , Endotelio Vascular/fisiopatología , Femenino , Humanos , Masculino , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/fisiología , Fumar/efectos adversos , Fumar/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Vasodilatación/fisiología , Adulto Joven
10.
Am J Respir Cell Mol Biol ; 48(1): 78-86, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23043086

RESUMEN

Although exposure to ambient hypoxia is known to cause proinflammatory vascular responses, the mechanisms initiating these responses are not understood. We tested the hypothesis that in systemic hypoxia, erythrocyte-derived H(2)O(2) induces proinflammatory gene transcription in vascular endothelium. We exposed mice or isolated, perfused murine lungs to 4 hours of hypoxia (8% O(2)). Leukocyte counts increased in the bronchoalveolar lavage. The expression of leukocyte adhesion receptors, reactive oxygen species, and protein tyrosine phosphorylation increased in freshly recovered lung endothelial cells (FLECs). These effects were inhibited by extracellular catalase and by the removal of erythrocytes, indicating that the responses were attributable to erythrocyte-derived H(2)O(2). Concomitant nuclear translocation of the p65 subunit of NF-κB and hypoxia-inducible factor-1α stabilization in FLECs occurred only in the presence of erythrocytes. Hemoglobin binding to the erythrocyte membrane protein, band 3, induced the release of H(2)O(2) from erythrocytes and the p65 translocation in FLECs. These data indicate for the first time, to our knowledge, that erythrocytes are responsible for endothelial transcriptional responses in hypoxia.


Asunto(s)
Eritrocitos/fisiología , Hipoxia/sangre , Hipoxia/fisiopatología , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Líquido del Lavado Bronquioalveolar/citología , Selectina E/sangre , Endotelio Vascular/fisiopatología , Peróxido de Hidrógeno/sangre , Hipoxia/genética , Mediadores de Inflamación/sangre , Pulmón/irrigación sanguínea , Pulmón/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1209-20, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22505671

RESUMEN

Although the lung expresses procoagulant proteins under inflammatory conditions, underlying mechanisms remain unclear. Here, we addressed lung endothelial expression of tissue factor (TF), which initiates the coagulation cascade and expression of which signifies development of a procoagulant phenotype in the vasculature. To establish the model of acid-induced acute lung injury (ALI), we intranasally instilled anesthetized mice with saline or acid. Then 2 h later, we isolated pulmonary vascular cells for flow cytometry and confocal microscopy to detect the leukocyte antigen, CD45 and the endothelial markers VE-cadherin and von Willebrand factor (vWf). Acid increased both the number of vWf-expressing cells as well as TF and P-selectin expressions on these cells. All of these effects were markedly inhibited by treating mice with antiplatelet serum, suggesting the involvement of platelets. The increased expressions of TF, vWf, and P-selectin in response to acid also occurred in platelets. Moreover, the effects were replicated in endothelial cells derived from isolated, blood-perfused lungs. However, the effect was inhibited completely in lungs perfused with platelet-depleted and, to a lesser extent, with leukocyte-depleted blood. Acid injury increased endothelial expressions of the platelet proteins, CD41 and CD42b, providing evidence that platelet proteins were transferred to the vascular surface. Reactive oxygen species (ROS) were implicated in these responses, in that the endothelial and platelet protein expressions were inhibited. We conclude that acid-induced ALI causes NOX2-mediated ROS generation that activates platelets, which then generate a procoagulant endothelial surface.


Asunto(s)
Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/inducido químicamente , Plaquetas/metabolismo , Endotelio Vascular/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tromboplastina/biosíntesis , Animales , Antígenos CD/biosíntesis , Coagulación Sanguínea , Plaquetas/inmunología , Cadherinas/biosíntesis , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Ácido Clorhídrico/efectos adversos , Ácido Clorhídrico/toxicidad , Antígenos Comunes de Leucocito/biosíntesis , Pulmón/inmunología , Pulmón/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Selectina-P/biosíntesis , Selectina-P/metabolismo , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/inmunología , Complejo GPIb-IX de Glicoproteína Plaquetaria/biosíntesis , Glicoproteína IIb de Membrana Plaquetaria/biosíntesis , Tromboplastina/metabolismo , Factor de von Willebrand/biosíntesis , Factor de von Willebrand/metabolismo
12.
Nat Med ; 18(5): 759-65, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22504485

RESUMEN

Bone marrow-derived stromal cells (BMSCs) protect against acute lung injury (ALI). To determine the role of BMSC mitochondria in this protection, we airway-instilled mice first with lipopolysaccharide (LPS) and then with either mouse BMSCs (mBMSCs) or human BMSCs (hBMSCs). Live optical studies revealed that the mBMSCs formed connexin 43 (Cx43)-containing gap junctional channels (GJCs) with the alveolar epithelia in these mice, releasing mitochondria-containing microvesicles that the epithelia engulfed. The presence of BMSC-derived mitochondria in the epithelia was evident optically, as well as by the presence of human mitochondrial DNA in mouse lungs instilled with hBMSCs. The mitochondrial transfer resulted in increased alveolar ATP concentrations. LPS-induced ALI, as indicated by alveolar leukocytosis and protein leak, inhibition of surfactant secretion and high mortality, was markedly abrogated by the instillation of wild-type mBMSCs but not of mutant, GJC-incompetent mBMSCs or mBMSCs with dysfunctional mitochondria. This is the first evidence, to our knowledge, that BMSCs protect against ALI by restituting alveolar bioenergetics through Cx43-dependent alveolar attachment and mitochondrial transfer.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Células de la Médula Ósea/fisiología , Mitocondrias/fisiología , Alveolos Pulmonares/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Conexina 43/fisiología , Metabolismo Energético , Uniones Comunicantes/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células del Estroma/fisiología
13.
J Clin Invest ; 121(5): 1986-99, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21519143

RESUMEN

Shedding of the extracellular domain of cytokine receptors allows the diffusion of soluble receptors into the extracellular space; these then bind and neutralize their cytokine ligands, thus dampening inflammatory responses. The molecular mechanisms that control this process, and the extent to which shedding regulates cytokine-induced microvascular inflammation, are not well defined. Here, we used real-time confocal microscopy of mouse lung microvascular endothelium to demonstrate that mitochondria are key regulators of this process. The proinflammatory cytokine soluble TNF-α (sTNF-α) increased mitochondrial Ca2+, and the purinergic receptor P2Y2 prolonged the response. Concomitantly, the proinflammatory receptor TNF-α receptor-1 (TNFR1) was shed from the endothelial surface. Inhibiting the mitochondrial Ca2+ increase blocked the shedding and augmented inflammation, as denoted by increases in endothelial expression of the leukocyte adhesion receptor E-selectin and in microvascular leukocyte recruitment. The shedding was also blocked in microvessels after knockdown of a complex III component and after mitochondria-targeted catalase overexpression. Endothelial deletion of the TNF-α converting enzyme (TACE) prevented the TNF-α receptor shedding response, which suggests that exposure of microvascular endothelium to sTNF-α induced a Ca2+-dependent increase of mitochondrial H2O2 that caused TNFR1 shedding through TACE activation. These findings provide what we believe to be the first evidence that endothelial mitochondria regulate TNFR1 shedding and thereby determine the severity of sTNF-α-induced microvascular inflammation.


Asunto(s)
Calcio/química , Pulmón/irrigación sanguínea , Mitocondrias/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Calcio/metabolismo , Selectina E/metabolismo , Humanos , Peróxido de Hidrógeno/química , Inflamación , Leucocitos/citología , Ratones , Ratones Endogámicos C57BL , Microcirculación , Modelos Biológicos , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...