Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Lab Invest ; 103(9): 100197, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37307952

RESUMEN

Acute respiratory distress syndrome (ARDS) is a leading cause of respiratory failure and death in patients in the intensive care unit. Experimentally, acute lung injury resolution depends on the repair of mitochondrial oxidant damage by the mitochondrial quality control (MQC) pathways, mitochondrial biogenesis, and mitophagy, but nothing is known about this in the human lung. In a case-control autopsy study, we compared the lungs of subjects dying of ARDS (n = 8; cases) and age-/gender-matched subjects dying of nonpulmonary causes (n = 7; controls). Slides were examined by light microscopy and immunofluorescence confocal microscopy, randomly probing for co-localization of citrate synthase with markers of oxidant stress, mitochondrial DNA damage, mitophagy, and mitochondrial biogenesis. ARDS lungs showed diffuse alveolar damage with edema, hyaline membranes, and neutrophils. Compared with controls, a high degree of mitochondrial oxidant damage was seen in type 2 epithelial (AT2) cells and alveolar macrophages by 8-hydroxydeoxyguanosine and malondialdehyde co-staining with citrate synthase. In ARDS, antioxidant protein heme oxygenase-1 and DNA repair enzyme N-glycosylase/DNA lyase (Ogg1) were found in alveolar macrophages but not in AT2 cells. Moreover, MAP1 light chain-3 (LC3) and serine/threonine-protein kinase (Pink1) staining were absent in AT2 cells, suggesting a mitophagy failure. Nuclear respiratory factor-1 staining was missing in the alveolar region, suggesting impaired mitochondrial biogenesis. Widespread hyperproliferation of AT2 cells in ARDS could suggest defective differentiation into type 1 cells. ARDS lungs show profuse mitochondrial oxidant DNA damage but little evidence of MQC activity in AT2 epithelium. Because these pathways are important for acute lung injury resolution, our findings support MQC as a novel pharmacologic target for ARDS resolution.


Asunto(s)
Lesión Pulmonar Aguda , Síndrome de Dificultad Respiratoria , Humanos , Citrato (si)-Sintasa/metabolismo , Pulmón/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Lesión Pulmonar Aguda/metabolismo , Oxidantes/metabolismo , Oxidantes/farmacología
2.
Exerc Sport Sci Rev ; 50(1): 49-55, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34690283

RESUMEN

Inducible heme oxygenase (HO)-1 catalyzes the breakdown of heme to biliverdin, iron, and carbon monoxide (CO). CO binds to cytochrome c oxidase and alters mitochondrial redox balance and coordinately regulates mitochondrial quality control (MQC) during oxidant stress and inflammation. The hypothesis presented is that the skeletal muscle HO-1/CO system helps modulate components in the MQC cycle during metabolic stress.


Asunto(s)
Monóxido de Carbono , Músculo Esquelético , Humanos , Inflamación , Estrés Fisiológico
3.
J Immunol ; 204(6): 1474-1485, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31996456

RESUMEN

An increasing body of evidence suggests that bone marrow-derived myeloid cells play a critical role in the pathophysiology of pulmonary hypertension (PH). However, the true requirement for myeloid cells in PH development has not been demonstrated, and a specific disease-promoting myeloid cell population has not been identified. Using bone marrow chimeras, lineage labeling, and proliferation studies, we determined that, in murine hypoxia-induced PH, Ly6Clo nonclassical monocytes are recruited to small pulmonary arteries and differentiate into pulmonary interstitial macrophages. Accumulation of these nonclassical monocyte-derived pulmonary interstitial macrophages around pulmonary vasculature is associated with increased muscularization of small pulmonary arteries and disease severity. To determine if the sensing of hypoxia by nonclassical monocytes contributes to the development of PH, mice lacking expression of hypoxia-inducible factor-1α in the Ly6Clo monocyte lineage were exposed to hypoxia. In these mice, vascular remodeling and PH severity were significantly reduced. Transcriptome analyses suggest that the Ly6Clo monocyte lineage regulates PH through complement, phagocytosis, Ag presentation, and chemokine/cytokine pathways. Consistent with these murine findings, relative to controls, lungs from pulmonary arterial hypertension patients displayed a significant increase in the frequency of nonclassical monocytes. Taken together, these findings show that, in response to hypoxia, nonclassical monocytes in the lung sense hypoxia, infiltrate small pulmonary arteries, and promote vascular remodeling and development of PH. Our results demonstrate that myeloid cells, specifically cells of the nonclassical monocyte lineage, play a direct role in the pathogenesis of PH.


Asunto(s)
Hipertensión Pulmonar/inmunología , Hipoxia/complicaciones , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Remodelación Vascular/inmunología , Animales , Antígenos Ly/metabolismo , Trasplante de Médula Ósea , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Humanos , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/cirugía , Hipoxia/inmunología , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/patología , Trasplante de Pulmón , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Quimera por Trasplante/inmunología , Remodelación Vascular/genética
4.
Proc Natl Acad Sci U S A ; 116(5): 1603-1612, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30655340

RESUMEN

Current therapeutic interventions for the treatment of respiratory infections are hampered by the evolution of multidrug resistance in pathogens as well as the lack of effective cellular targets. Despite the identification of multiple region-specific lung progenitor cells, the identity of molecules that might be therapeutically targeted in response to infections to promote activation of progenitor cell types remains elusive. Here, we report that loss of Abl1 specifically in SCGB1A1-expressing cells leads to a significant increase in the proliferation and differentiation of bronchiolar epithelial cells, resulting in dramatic expansion of an SCGB1A1+ airway cell population that coexpresses SPC, a marker for type II alveolar cells that promotes alveolar regeneration following bacterial pneumonia. Furthermore, treatment with an Abl-specific allosteric inhibitor enhanced regeneration of the alveolar epithelium and promoted accelerated recovery of mice following pneumonia. These data reveal a potential actionable target that may be exploited for efficient recovery after pathogen-induced infections.


Asunto(s)
Pulmón/metabolismo , Pulmón/fisiopatología , Neumonía Bacteriana/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Regeneración/fisiología , Células Madre/metabolismo , Uteroglobina/metabolismo , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/fisiología , Animales , Bronquiolos/metabolismo , Bronquiolos/fisiopatología , Diferenciación Celular/fisiología , Línea Celular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía Bacteriana/fisiopatología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/fisiopatología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/fisiopatología , Células Madre/fisiología
5.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L321-L335, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34105359

RESUMEN

Bacterial pneumonia is a major cause of morbidity and mortality worldwide despite the use of antibiotics, and novel therapies are urgently needed. Building on previous work, we aimed to 1) develop a baboon model of severe pneumococcal pneumonia and sepsis with organ dysfunction and 2) test the safety and efficacy of a novel extracorporeal blood filter to remove proinflammatory molecules and improve organ function. After a dose-finding pilot study, 12 animals were inoculated with Streptococcus pneumoniae [5 × 109 colony-forming units (CFU)], given ceftriaxone at 24 h after inoculation, and randomized to extracorporeal blood purification using a filter coated with surface-immobilized heparin sulfate (n = 6) or sham treatment (n = 6) for 4 h at 30 h after inoculation. For safety analysis, four uninfected animals also underwent purification. At 48 h, necropsy was performed. Inoculated animals developed severe pneumonia and septic shock. Compared with sham-treated animals, septic animals treated with purification displayed significantly less kidney injury, metabolic acidosis, hypoglycemia, and shock (P < 0.05). Purification blocked the rise in peripheral blood S. pneumoniae DNA, attenuated bronchoalveolar lavage (BAL) CCL4, CCL2, and IL-18 levels, and reduced renal oxidative injury and classical NLRP3 inflammasome activation. Purification was safe in both uninfected and infected animals and produced no adverse effects. We demonstrate that heparin-based blood purification significantly attenuates levels of circulating S. pneumoniae DNA and BAL cytokines and is renal protective in baboons with severe pneumococcal pneumonia and septic shock. Purification was associated with less severe acute kidney injury, metabolic derangements, and shock. These results support future clinical studies in critically ill septic patients.


Asunto(s)
Hemofiltración , Heparina/química , Neumonía Neumocócica/terapia , Choque Séptico/terapia , Streptococcus pneumoniae/metabolismo , Animales , Citocinas/metabolismo , Masculino , Papio , Proyectos Piloto , Neumonía Neumocócica/sangre , Choque Séptico/sangre
6.
Annu Rev Physiol ; 79: 495-515, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-27959621

RESUMEN

Remarkable new roles for mitochondria in calcium handling, apoptosis, heme turnover, inflammation, and oxygen and nutrient sensing have been discovered for organelles that were once thought to be simple energy converters. Although deficits in mitochondrial function are often associated with energy failure and apoptosis, working cells maintain a mitochondrial reserve that affords the organelles distinct homeostatic sensing and regulatory abilities in lung cells. As primary intracellular sources of oxidants, mitochondria serve as critical monitors and modulators of vital oxidation-reduction processes, including mitochondrial biogenesis, mitophagy, inflammasome activation, cell proliferation, and prevention of fibrosis. These processes participate in disease pathogenesis in all lung regions mainly when interference with mitochondrial quality control mechanisms impedes their roles in maintenance of lung health. Sharper identification of mitochondrial-driven signaling mechanisms in specific lung cell types will better refine our understanding of respiratory disease pathogenesis and lead to new diagnostic and therapeutic measures to support mitochondrial quality.


Asunto(s)
Enfermedades Pulmonares/patología , Pulmón/patología , Mitocondrias/patología , Animales , Humanos , Inflamación/patología , Transducción de Señal/fisiología
7.
Am J Physiol Cell Physiol ; 319(4): C746-C756, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32845721

RESUMEN

Nutrient excess increases skeletal muscle oxidant production and mitochondrial fragmentation that may result in impaired mitochondrial function, a hallmark of skeletal muscle insulin resistance. This led us to explore whether an endogenous gas molecule, carbon monoxide (CO), which is thought to prevent weight gain and metabolic dysfunction in mice consuming high-fat diets, alters mitochondrial morphology and respiration in C2C12 myoblasts exposed to high glucose (15.6 mM) and high fat (250 µM BSA-palmitate) (HGHF). Also, skeletal muscle mitochondrial morphology, distribution, respiration, and energy expenditure were examined in obese resistant (OR) and obese prone (OP) rats that consumed a high-fat and high-sucrose diet for 10 wk with or without intermittent low-dose inhaled CO and/or exercise training. In cells exposed to HGHF, superoxide production, mitochondrial membrane potential (ΔΨm), mitochondrial fission regulatory protein dynamin-related protein 1 (Drp1) and mitochondrial fragmentation increased, while mitochondrial respiratory capacity was reduced. CO decreased HGHF-induced superoxide production, Drp1 protein levels and mitochondrial fragmentation, maintained ΔΨm, and increased mitochondrial respiratory capacity. In comparison with lean OR rats, OP rats had smaller skeletal muscle mitochondria that contained disorganized cristae, a normal mitochondrial distribution, but reduced citrate synthase protein expression, normal respiratory responses, and a lower energy expenditure. The combination of inhaled CO and exercise produced the greatest effect on mitochondrial morphology, increasing ADP-stimulated respiration in the presence of pyruvate, and preventing a decline in resting energy expenditure. These data support a therapeutic role for CO and exercise in preserving mitochondrial morphology and respiration during metabolic overload.


Asunto(s)
Monóxido de Carbono/metabolismo , Dinaminas/genética , Obesidad/genética , Aumento de Peso/genética , Animales , Monóxido de Carbono/farmacología , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Humanos , Ratones , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/patología , Dinámicas Mitocondriales/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mioblastos/metabolismo , Mioblastos/patología , Obesidad/metabolismo , Obesidad/patología , Condicionamiento Físico Animal , Ratas , Especies Reactivas de Oxígeno/metabolismo , Sacarosa/efectos adversos
8.
Circulation ; 139(13): 1629-1642, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30586762

RESUMEN

BACKGROUND: Receptor signaling is central to vascular endothelial function and is dysregulated in vascular diseases such as atherosclerosis and pulmonary arterial hypertension (PAH). Signaling pathways involved in endothelial function include vascular endothelial growth factor receptors (VEGFRs) and G protein-coupled receptors, which classically activate distinct intracellular signaling pathways and responses. The mechanisms that regulate these signaling pathways have not been fully elucidated and it is unclear what nodes for cross talk exist between these diverse signaling pathways. For example, multifunctional ß-arrestin (ARRB) adapter proteins are best known as regulators of G protein-coupled receptor signaling, but their role at other receptors and their physiological importance in the setting of vascular disease are unclear. METHODS: We used a combination of human samples from PAH, human microvascular endothelial cells from lung, and Arrb knockout mice to determine the role of ARRB1 in endothelial VEGFR3 signaling. In addition, a number of biochemical analyses were performed to determine the interaction between ARRB1 and VEGFR3, signaling mediators downstream of VEGFR3, and the internalization of VEGFR3. RESULTS: Expression of ARRB1 and VEGFR3 was reduced in human PAH, and the deletion of Arrb1 in mice exposed to hypoxia led to worse PAH with a loss of VEGFR3 signaling. Knockdown of ARRB1 inhibited VEGF-C-induced endothelial cell proliferation, migration, and tube formation, along with reduced VEGFR3, Akt, and endothelial nitric oxide synthase phosphorylation. This regulation was mediated by direct ARRB1 binding to the VEGFR3 kinase domain and resulted in decreased VEGFR3 internalization. CONCLUSIONS: Our results demonstrate a novel role for ARRB1 in VEGFR regulation and suggest a mechanism for cross talk between G protein-coupled receptors and VEGFRs in PAH. These findings also suggest that strategies to promote ARRB1-mediated VEGFR3 signaling could be useful in the treatment of pulmonary hypertension and other vascular disease.


Asunto(s)
Endotelio Vascular/metabolismo , Hipertensión Pulmonar/metabolismo , Transducción de Señal , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , beta-Arrestina 1/metabolismo , Animales , Endotelio Vascular/patología , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Masculino , Ratones , Ratones Noqueados , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , beta-Arrestina 1/genética
9.
Crit Care Med ; 47(5): 651-658, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30730439

RESUMEN

OBJECTIVES: Metabolic derangements in sepsis stem from mitochondrial injury and contribute significantly to organ failure and mortality; however, little is known about mitochondrial recovery in human sepsis. We sought to test markers of mitochondrial injury and recovery (mitochondrial biogenesis) noninvasively in peripheral blood mononuclear cells from patients with sepsis and correlate serial measurements with clinical outcomes. DESIGN: Prospective case-control study. SETTING: Academic Medical Center and Veterans Affairs Hospital. PATIENTS: Uninfected control patients (n = 20) and septic ICU patients (n = 37). INTERVENTIONS: Blood samples were collected once from control patients and serially with clinical data on days 1, 3, and 5 from septic patients. Gene products for HMOX1, NRF1, PPARGC1A, and TFAM, and mitochondrial DNA ND1 and D-loop were measured by quantitative reverse transcriptase-polymerase chain reaction. Proinflammatory cytokines were measured in plasma and neutrophil lysates. MEASUREMENTS AND MAIN RESULTS: Median (interquartile range) Acute Physiology and Chronic Health Evaluation II and Sequential Organ Failure Assessment scores were 21 (8) and 10 (4), respectively, and 90-day mortality was 19%. Transcript levels of all four genes in peripheral blood mononuclear cells were significantly reduced in septic patients on day 1 (p < 0.05), whereas mitochondrial DNA copy number fell and plasma D-loop increased (both p < 0.05), indicative of mitochondrial damage. D-loop content was directly proportional to tumor necrosis factor-α and high-mobility group protein B1 cytokine expression. By day 5, we observed transcriptional activation of mitochondrial biogenesis and restoration of mitochondrial DNA copy number (p < 0.05). Patients with early activation of mitochondrial biogenesis were ICU-free by 1 week. CONCLUSIONS: Our findings support data that sepsis-induced mitochondrial damage is reversed by activation of mitochondrial biogenesis and that gene transcripts measured noninvasively in peripheral blood mononuclear cells can serve as novel biomarkers of sepsis recovery.


Asunto(s)
ADN Mitocondrial/sangre , Leucocitos Mononucleares/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Sepsis/metabolismo , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/genética , Enfermedades Mitocondriales/sangre , Enfermedades Mitocondriales/genética , Estudios Prospectivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Sepsis/sangre , Sepsis/genética
10.
Exp Cell Res ; 363(2): 151-159, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29291399

RESUMEN

Microparticles (MPs) are small membrane-bound vesicles released from activated or dying cells. As shown previously, LPS stimulation of the RAW 264.7 macrophage cell line can induce MP release, with the caspase inhibitor Z-VAD increasing the extent of this process. Since combined treatment of cells with LPS and Z-VAD can induce necroptosis, we explored particle release during this form of cell death using flow cytometry to assess particle size, binding of annexin V and staining for DNA with propidium iodide (PI) and SYTO 13. The role of necroptosis was assessed by determining the effects of necrostatin, an inhibitor of RIP1, a kinase regulating this form of cell death. These studies demonstrated that, during necroptosis, RAW 264.7 cells release MPs that resemble those released from cells treated with staurosporine to induce apoptosis. The particles contained DNA as determined by binding of PI and SYTO 13, with PCR analysis demonstrating both chromosomal and mitochondrial DNA. The presence of mitochondria in the MP preparations was demonstrated by staining with MitoTracker Green. Flow cytometry indicated that purified mitochondria have properties of MPs. Together, these studies indicate that cells undergoing necroptosis can release MPs and that mitochondria can be components of MP preparations.


Asunto(s)
Muerte Celular/efectos de los fármacos , Micropartículas Derivadas de Células/efectos de los fármacos , Micropartículas Derivadas de Células/metabolismo , Macrófagos/metabolismo , Mitocondrias/metabolismo , Animales , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Macrófagos/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Necrosis/metabolismo , Células RAW 264.7/metabolismo , Estaurosporina/farmacología
11.
Pharmacol Rev ; 68(1): 20-48, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26589414

RESUMEN

In addition to oxidative phosphorylation (OXPHOS), mitochondria perform other functions such as heme biosynthesis and oxygen sensing and mediate calcium homeostasis, cell growth, and cell death. They participate in cell communication and regulation of inflammation and are important considerations in aging, drug toxicity, and pathogenesis. The cell's capacity to maintain its mitochondria involves intramitochondrial processes, such as heme and protein turnover, and those involving entire organelles, such as fusion, fission, selective mitochondrial macroautophagy (mitophagy), and mitochondrial biogenesis. The integration of these processes exemplifies mitochondrial quality control (QC), which is also important in cellular disorders ranging from primary mitochondrial genetic diseases to those that involve mitochondria secondarily, such as neurodegenerative, cardiovascular, inflammatory, and metabolic syndromes. Consequently, mitochondrial biology represents a potentially useful, but relatively unexploited area of therapeutic innovation. In patients with genetic OXPHOS disorders, the largest group of inborn errors of metabolism, effective therapies, apart from symptomatic and nutritional measures, are largely lacking. Moreover, the genetic and biochemical heterogeneity of these states is remarkably similar to those of certain acquired diseases characterized by metabolic and oxidative stress and displaying wide variability. This biologic variability reflects cell-specific and repair processes that complicate rational pharmacological approaches to both primary and secondary mitochondrial disorders. However, emerging concepts of mitochondrial turnover and dynamics along with new mitochondrial disease models are providing opportunities to develop and evaluate mitochondrial QC-based therapies. The goals of such therapies extend beyond amelioration of energy insufficiency and tissue loss and entail cell repair, cell replacement, and the prevention of fibrosis. This review summarizes current concepts of mitochondria as disease elements and outlines novel strategies to address mitochondrial dysfunction through the stimulation of mitochondrial biogenesis and quality control.


Asunto(s)
Mitocondrias/metabolismo , Enfermedades Mitocondriales/fisiopatología , Monóxido de Carbono/metabolismo , Eritropoyetina/metabolismo , Estrógenos/metabolismo , Depuradores de Radicales Libres/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Enfermedades Mitocondriales/inducido químicamente , Mitofagia/fisiología , Óxido Nítrico/metabolismo , Fosforilación Oxidativa , Polifenoles/metabolismo , Hormonas Tiroideas/metabolismo
12.
Nitric Oxide ; 74: 56-64, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29355776

RESUMEN

S-nitrosothiols derived from nitric oxide are known to regulate cell signaling through thiol modification. Since small G protein RhoA contains cysteine residues in the GTP-binding domain which is critical for its function, modification these thiols may alter RhoA activity and lead to changes in the downstream signaling such as myosin light chain phosphorylation. However, it is still unclear that if RhoA activity and its downstream signals might be modulated by S-nitrosothiols and if the two cysteine residues located in the GTP-binding domain are critical for the regulation. In this study we show that S-nitroso-L-cysteine (CSNO) blocked RhoA activation as determined by either GDP/GTP exchange, active RhoA binding to rhotekin or RhoA translocation. CSNO was shown to lead to RhoA nitrosylation and RhoA thiol oxidation status was found to be consistent with loss of its activity. Mutation of all 6 single cysteine residues to serine showed that purified recombinant C20S mutant and C26/20S mutant were resistant to CSNO, but interestingly, in the intact cells only the double C16/20S mutant was resistant to CSNO. Moreover, inhibition of RhoA activation led to Rho-kinase inhibition and inhibition of Rho pathway signaling by CSNO. In both smooth muscle cells and aortic tissue, the outcome was inhibition of agonist-stimulated MYPT1 phosphorylation and reduced levels of myosin light chain phosphorylation. These effects of CSNO on MYPT1 and myosin light chain phosphorylation appear to be cGMP-independent since they were unaffected by inhibition of guanylyl cyclase. In contrast to CSNO, spermine NONOate did not alter RhoA GDP/GTP exchange and the effects of this compound on myosin light chain phosphorylation were blocked by guanylyl cyclase inhibition. And importantly, in C16/20S overexpressed smooth muscle cells, MYPT1 phosphorylation was resistant to the inhibitory effect of CSNO. Together, these data suggest that S-nitrosothiols regulate myosin light chain phosphorylation by inhibiting RhoA/Rho-kinase signaling through modification of RhoA cysteine residues at 16 and 20 in its GTP-binding domain, which might be an important therapeutic target for diseases with imbalanced vascular resistance.


Asunto(s)
Contracción Muscular , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , S-Nitrosotioles/metabolismo , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo , Animales , Células HEK293 , Humanos , Ratas
13.
Am J Respir Cell Mol Biol ; 56(3): 300-309, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27636016

RESUMEN

Oxidative stress resulting from inflammatory responses that occur during acute lung injury and sepsis can initiate changes in mitochondrial function. Autophagy regulates cellular processes in the setting of acute lung injury, sepsis, and oxidative stress by modulating the immune response and facilitating turnover of damaged cellular components. We have shown that mesenchymal stromal cells (MSCs) improve survival in murine models of sepsis by also regulating the immune response. However, the effect of autophagy on MSCs and MSC mitochondrial function during oxidative stress is unknown. This study investigated the effect of depletion of autophagic protein microtubule-associated protein 1 light chain 3B (LC3B) and beclin 1 (BECN1) on the response of MSCs to oxidative stress. MSCs were isolated from wild-type (WT) and LC3B-/- or Becn1+/- mice. MSCs from the LC3B-/- and Becn1+/- animals had increased susceptibility to oxidative stress-induced cell death as compared with WT MSCs. The MSCs depleted of autophagic proteins also had impaired mitochondrial function (decreased intracellular ATP, reduced mitochondrial membrane potential, and increased mitochondrial reactive oxygen species production) under oxidative stress as compared with WT MSCs. In WT MSCs, carbon monoxide (CO) preconditioning enhanced autophagy and mitophagy, and rescued the cells from oxidative stress-induced death. CO preconditioning was not able to rescue the decreased survival of MSCs from the LC3B-/- and Becn1+/- animals, further supporting the tenet that CO exerts its cytoprotective effects via the autophagy pathway.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Mitocondrias/metabolismo , Estrés Oxidativo , Adenosina Trifosfato/metabolismo , Animales , Antioxidantes/metabolismo , Autofagia/efectos de los fármacos , Monóxido de Carbono/farmacología , Células Cultivadas , Espacio Intracelular/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fenotipo
14.
Am J Physiol Lung Cell Mol Physiol ; 313(4): L699-L709, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28663335

RESUMEN

Mitochondrial damage is often overlooked in acute lung injury (ALI), yet most of the lung's physiological processes, such as airway tone, mucociliary clearance, ventilation-perfusion (Va/Q) matching, and immune surveillance require aerobic energy provision. Because the cell's mitochondrial quality control (QC) process regulates the elimination and replacement of damaged mitochondria to maintain cell survival, we serially evaluated mitochondrial biogenesis and mitophagy in the alveolar regions of mice in a validated Staphylococcus aureus pneumonia model. We report that apart from cell lysis by direct contact with microbes, modest epithelial cell death was detected despite significant mitochondrial damage. Cell death by TdT-mediated dUTP nick-end labeling staining occurred on days 1 and 2 postinoculation: apoptosis shown by caspase-3 cleavage was present on days 1 and 2, while necroptosis shown by increased levels of phospho- mixed lineage kinase domain-like protein (MLKL) and receptor-interacting serine/threonine-protein kinase 1 (RIPK1) was present on day 1 Cell death in alveolar type I (AT1) cells assessed by bronchoalveolar lavage fluid receptor for advanced glycation end points (RAGE) levels was high, yet AT2 cell death was limited while both mitochondrial biogenesis and mitophagy were induced. These mitochondrial QC mechanisms were evaluated mainly in AT2 cells by localizing increases in citrate synthase content, increases in nuclear mitochondrial biogenesis regulators nuclear respiratory factor-1 (NRF-1) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), and increases in light chain 3B protein (LC3-I)/LC3II ratios. Concomitant changes in p62, Pink 1, and Parkin protein levels indicated activation of mitophagy. By confocal microscopy, mitochondrial biogenesis and mitophagy were often observed on day 1 within the same AT2 cells. These findings imply that mitochondrial QC activation in pneumonia-damaged AT2 cells promotes cell survival in support of alveolar function.


Asunto(s)
Células Epiteliales Alveolares/patología , Mitocondrias/patología , Neumonía Estafilocócica/etiología , Neumonía Estafilocócica/patología , Infecciones Estafilocócicas/complicaciones , Staphylococcus aureus/patogenicidad , Células Epiteliales Alveolares/metabolismo , Animales , Apoptosis , Biomarcadores/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neumonía Estafilocócica/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología
15.
Can J Physiol Pharmacol ; 95(9): 1009-1018, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28467859

RESUMEN

Hypoxemia can be life-threatening, both acutely and chronically. Because hypoxemia causes vascular dysregulation that further restricts oxygen availability to tissue, it can be pharmacologically addressed. We hypothesized that theophylline can be safely combined with the ß2-adrenergic vasodilator bambuterol to improve oxygen availability in hypoxemic patients. Ergogenicity and hemodynamic effects of bambuterol and theophylline were measured in rats under hypobaric and normobaric hypoxia (12% O2). Feasibility in humans was assessed using randomized, double-blind testing of the influence of combined slow-release theophylline (300 mg) and bambuterol (20 mg) on adverse events (AEs), plasma K+, pulse, blood pressure, and drug interaction. Both drugs and their combination significantly improved hypoxic endurance in rats. In humans, common AEs were low K+ (<3.5 mmol/L; bambuterol: 12, theophylline: 4, combination: 13 episodes) and tremors (10, 0, 14 episodes). No exacerbation or serious AE occurred when drugs were combined. A drop in plasma K+ coincided with peak bambuterol plasma concentrations. Bambuterol increased heart rate by approximately 13 bpm. Drug interaction was present but small. We report promise, feasibility, and relative safety of combined theophylline and bambuterol as a treatment of hypoxemia in humans. Cardiac safety and blood K+ will be important safety endpoints when testing these drugs in hypoxemic subjects.


Asunto(s)
Hipoxia/tratamiento farmacológico , Terbutalina/análogos & derivados , Teofilina/farmacología , Adulto , Animales , Disponibilidad Biológica , Interacciones Farmacológicas , Femenino , Semivida , Hemodinámica/efectos de los fármacos , Humanos , Hipoxia/sangre , Hipoxia/fisiopatología , Masculino , Condicionamiento Físico Animal , Ratas , Seguridad , Terbutalina/efectos adversos , Terbutalina/farmacocinética , Terbutalina/farmacología , Terbutalina/uso terapéutico , Teofilina/efectos adversos , Teofilina/farmacocinética , Teofilina/uso terapéutico , Resultado del Tratamiento , Adulto Joven
16.
Am J Respir Crit Care Med ; 193(5): 552-60, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26488115

RESUMEN

RATIONALE: Central airway stenosis (CAS) after lung transplantation has been attributed in part to chronic airway ischemia; however, little is known about the time course or significance of large airway hypoxia early after transplantation. OBJECTIVES: To evaluate large airway oxygenation and hypoxic gene expression during the first month after lung transplantation and their relation to airway complications. METHODS: Subjects who underwent lung transplantation underwent endobronchial tissue oximetry of native and donor bronchi at 0, 3, and 30 days after transplantation (n = 11) and/or endobronchial biopsies (n = 14) at 30 days for real-time polymerase chain reaction of hypoxia-inducible genes. Patients were monitored for 6 months for the development of transplant-related complications. MEASUREMENTS AND MAIN RESULTS: Compared with native endobronchial tissues, donor tissue oxygen saturations (Sto2) were reduced in the upper lobes (74.1 ± 1.8% vs. 68.8 ± 1.7%; P < 0.05) and lower lobes (75.6 ± 1.6% vs. 71.5 ± 1.8%; P = 0.065) at 30 days post-transplantation. Donor upper lobe and subcarina Sto2 levels were also lower than the main carina (difference of -3.9 ± 1.5 and -4.8 ± 2.1, respectively; P < 0.05) at 30 days. Up-regulation of hypoxia-inducible genes VEGFA, FLT1, VEGFC, HMOX1, and TIE2 was significant in donor airways relative to native airways (all P < 0.05). VEGFA, KDR, and HMOX1 were associated with prolonged respiratory failure, prolonged hospitalization, extensive airway necrosis, and CAS (P < 0.05). CONCLUSIONS: These findings implicate donor bronchial hypoxia as a driving factor for post-transplantation airway complications. Strategies to improve airway oxygenation, such as bronchial artery re-anastomosis and hyperbaric oxygen therapy merit clinical investigation.


Asunto(s)
Bronquios/metabolismo , Hipoxia de la Célula/genética , Enfermedades Pulmonares/cirugía , Trasplante de Pulmón , Complicaciones Posoperatorias/genética , Insuficiencia Respiratoria/genética , Trasplantes/metabolismo , Adulto , Anciano , Bronquios/irrigación sanguínea , Bronquios/patología , Constricción Patológica/genética , Fibrosis Quística/cirugía , Femenino , Expresión Génica , Hemo-Oxigenasa 1/genética , Humanos , Fibrosis Pulmonar Idiopática/cirugía , Tiempo de Internación , Enfermedades Pulmonares Intersticiales/cirugía , Masculino , Persona de Mediana Edad , Necrosis/genética , Oximetría , Pronóstico , Enfermedad Pulmonar Obstructiva Crónica/cirugía , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor TIE-2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoidosis Pulmonar/cirugía , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
17.
Clin Transplant ; 30(9): 1134-9, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27410718

RESUMEN

BACKGROUND: Central airway stenosis (CAS) is common after lung transplantation and causes significant post-transplant morbidity. It is often preceded by extensive airway necrosis, related to airway ischemia. Hyperbaric oxygen therapy (HBOT) is useful for ischemic grafts and may reduce the development of CAS. METHODS: The purpose of this study was to determine whether HBOT could be safely administered to lung transplant patients with extensive necrotic airway plaques. Secondarily, we assessed any effects of HBOT on the incidence and severity of CAS. Patients with extensive necrotic airway plaques within 1-2 months after lung transplantation were treated with HBOT along with standard care. These patients were compared with a contemporaneous reference group with similar plaques who did not receive HBOT. RESULTS: Ten patients received HBOT for 18.5 (interquartile range, IQR 11-20) sessions, starting at 40.5 (IQR 34-54) days after transplantation. HBOT was well tolerated. Incidence of CAS was similar between HBOT-treated patients and reference patients (70% vs 87%, respectively; P=.34), but fewer stents were required in HBOT patients (10% vs 56%, respectively; P=.03). CONCLUSIONS: This pilot study is the first to demonstrate HBOT safety in patients who develop necrotic airway plaques after lung transplantation. HBOT may reduce the need for airway stent placement in patients with CAS.


Asunto(s)
Obstrucción de las Vías Aéreas/terapia , Oxigenoterapia Hiperbárica/métodos , Trasplante de Pulmón/efectos adversos , Complicaciones Posoperatorias/terapia , Obstrucción de las Vías Aéreas/epidemiología , Obstrucción de las Vías Aéreas/etiología , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Masculino , Persona de Mediana Edad , North Carolina/epidemiología , Proyectos Piloto , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Factores de Tiempo , Resultado del Tratamiento
18.
Proc Natl Acad Sci U S A ; 110(28): 11529-34, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23798386

RESUMEN

From the perspectives of disease transmission and sterility maintenance, the world's blood supplies are generally safe. However, in multiple clinical settings, red blood cell (RBC) transfusions are associated with adverse cardiovascular events and multiorgan injury. Because ∼85 million units of blood are administered worldwide each year, transfusion-related morbidity and mortality is a major public health concern. Blood undergoes multiple biochemical changes during storage, but the relevance of these changes to unfavorable outcomes is unclear. Banked blood shows reduced levels of S-nitrosohemoglobin (SNO-Hb), which in turn impairs the ability of stored RBCs to effect hypoxic vasodilation. We therefore reasoned that transfusion of SNO-Hb-deficient blood may exacerbate, rather than correct, impairments in tissue oxygenation, and that restoration of SNO-Hb levels would improve transfusion efficacy. Notably in mice, administration of banked RBCs decreased skeletal muscle pO2, but infusion of renitrosylated cells maintained tissue oxygenation. In rats, hemorrhage-induced reductions in muscle pO2 were corrected by SNO-Hb-repleted RBCs, but not by control, stored RBCs. In anemic awake sheep, stored renitrosylated, but not control RBCs, produced sustained improvements in O2 delivery; in anesthetized sheep, decrements in hemodynamic status, renal blood flow, and kidney function incurred following transfusion of banked blood were also prevented by renitrosylation. Collectively, our findings lend support to the idea that transfusions may be causally linked to ischemic events and suggest a simple approach to prevention (i.e., SNO-Hb repletion). If these data are replicated in clinical trials, renitrosylation therapy could have significant therapeutic impact on the care of millions of patients.


Asunto(s)
Transfusión Sanguínea , Compuestos Nitrosos/metabolismo , Oxígeno/metabolismo , Anemia/terapia , Animales , Hemorragia/terapia , Ratones , Ratas , Ovinos
19.
Am J Respir Cell Mol Biol ; 53(3): 314-25, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25568926

RESUMEN

Strategies for the treatment of bacterial pneumonia beyond traditional antimicrobial therapy have been limited. The recently discovered novel genus of lipid mediators, coined "specialized proresolving mediators" (SPMs), which orchestrate clearance of recruited leukocytes and restore epithelial barrier integrity, have offered new insight into the resolution of inflammation. We performed lipid mediator (LM) metabololipidomic profiling and identification of LMs on peripheral blood leukocytes and plasma from a baboon model of Streptococcus pneumoniae pneumonia. Leukocytes and plasma were isolated from whole blood of S. pneumoniae-infected (n = 5-6 per time point) and control, uninfected baboons (n = 4 per time point) at 0, 24, 48, and 168 hours. In a subset of baboons with pneumonia (n = 3), we administered inhaled carbon monoxide (CO) at 48 hours (200-300 ppm for 60-90 min). Unstimulated leukocytes from control animals produced a proresolving LM signature with elevated resolvins and lipoxins. In contrast, serum-treated, zymosan-stimulated leukocytes and leukocytes from baboons with S. pneumoniae pneumonia produced a proinflammatory LM signature profile with elevated leukotriene B4 and prostaglandins. Plasma from baboons with S. pneumoniae pneumonia also displayed significantly reduced LM-SPM levels, including eicosapentaenoic acid-derived E-series resolvins (RvE) and lipoxins. CO inhalation increased levels of plasma RvE and lipoxins relative to preexposure levels. These results establish the leukocyte and plasma LM profiles biosynthesized during S. pneumoniae pneumonia in baboons and provide evidence for pneumonia-induced dysregulation of these proresolution programs. Moreover, these SPM profiles are partially restored with inhaled low-dose CO and SPM, which may shorten the time to pneumonia resolution.


Asunto(s)
Monóxido de Carbono/administración & dosificación , Factores Inmunológicos/administración & dosificación , Lípidos/sangre , Neumonía Neumocócica/sangre , Administración por Inhalación , Animales , Evaluación Preclínica de Medicamentos , Leucocitos Mononucleares/metabolismo , Leucotrieno B4/sangre , Metabolismo de los Lípidos , Masculino , Metaboloma , Papio , Neumonía Neumocócica/tratamiento farmacológico , Neumonía Neumocócica/inmunología , Streptococcus pneumoniae/inmunología
20.
J Biol Chem ; 289(1): 41-52, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24253037

RESUMEN

A key transcriptional regulator of cell metabolism, the peroxisome proliferator-activated receptor γ co-activator 1-α (PPARGC-1-α or PGC-1α), also regulates mitochondrial biogenesis, but its role in antioxidant gene regulation is not well understood. Here, we asked whether genetic heterozygosity of PGC-1α modulates gene expression for the mitochondrial antioxidant enzyme SOD-2 during hepatic inflammatory stress. Using Staphylococcus aureus peritonitis in mice, we found significant Sod2 gene induction in WT mice, whereas PGC-1α heterozygotes (PGC-1α(+/-)) failed to augment Sod2 mRNA and protein levels. Impaired Sod2 regulation in PGC-1α(+/-) mice was accompanied by oxidative stress shown by elevated mitochondrial GSSG/GSH and protein carbonyls. In silico analysis of the mouse proximal Sod2 promoter region revealed consensus binding sites for the Nfe2l2 (Nrf2) transcription factor. Chromatin immunoprecipitation demonstrated diminished Nfe2l2 protein binding to the antioxidant response element promoter site proximal to the Sod2 start site in PGC-1α heterozygous mice, implicating PGC-1α in facilitation of Nfe2l2 DNA binding. Nuclear protein co-immunoprecipitation demonstrated an interaction between hepatic Nfe2l2 and PGC-1α in WT mice that was greatly reduced in PGC-1α(+/-) mice. The data indicate that PGC-1α promotes mitochondrial antioxidant enzyme expression through Nfe2l2-mediated SOD-2 expression in sepsis. The presence of this new PGC-1α-dependent signaling axis indicates that PGC-1α opposes mitochondrial oxidative stress by means of selective induction of one or more antioxidant response element-driven genes. By implication, exploitation of this axis could lead to new pharmacological interventions to improve the antioxidant defenses during oxidative stress-induced mitochondrial damage.


Asunto(s)
Hígado/metabolismo , Mitocondrias Hepáticas/metabolismo , Estrés Oxidativo , Sepsis/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus , Factores de Transcripción/metabolismo , Animales , Regulación Enzimológica de la Expresión Génica/genética , Hígado/patología , Ratones , Ratones Mutantes , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/patología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Peritonitis/genética , Peritonitis/metabolismo , Peritonitis/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Elementos de Respuesta/genética , Sepsis/genética , Sepsis/patología , Transducción de Señal/genética , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/patología , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA