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












Base de datos
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.
iScience ; 25(1): 103535, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-34977500

RESUMEN

The preclinical model of bleomycin-induced lung fibrosis is useful to study mechanisms related to human pulmonary fibrosis. Using BLM in mice, we find low HO-1 expression. Although a unique Rhenium-CO-releasing molecule (ReCORM) up-regulates HO-1, NRF-1, CCN5, and SMAD7, it reduces TGFß1, TGFßr1, collagen, α-SMA, and phosphorylated Smad2/3 levels in mouse lung and in human lung fibroblasts. ChIP assay studies confirm NRF-1 binding to the promoters of TGFß1 repressors CCN5 and Smad7. ReCORM did not blunt lung fibrosis in Hmox1-deficient alveolar type 2 cell knockout mice, suggesting this gene participates in lung protection. In human lung fibroblasts, TGFß1-dependent production of α-SMA is abolished by ReCORM or by NRF-1 gene transfection. We demonstrate effective HO-1/NRF-1 signaling in lung AT2 cells protects against BLM induced lung injury and fibrosis by maintaining mitochondrial health, function, and suppressing the TGFß1 pathway. Thus, protection of AT2 cell mitochondrial integrity via HO-1/NRF-1 presents an innovative therapeutic target.

3.
Front Mol Neurosci ; 15: 1062410, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36704328

RESUMEN

Oxygen breathing at elevated partial pressures (PO2's) at or more than 3 atmospheres absolute (ATA) causes a reduction in brain γ-aminobutyric acid (GABA) levels that impacts the development of central nervous system oxygen toxicity (CNS-OT). Drugs that increase brain GABA content delay the onset of CNS-OT, but it is unknown if oxidant damage is lessened because brain tissue PO2 remains elevated during hyperbaric oxygen (HBO2) exposures. Experiments were performed in rats and mice to measure brain GABA levels with or without GABA transporter inhibitors (GATs) and its influence on cerebral blood flow, oxidant damage, and aspects of mitochondrial quality control signaling (mitophagy and biogenesis). In rats pretreated with tiagabine (GAT1 inhibitor), the tachycardia, secondary rise in mean arterial blood pressure, and cerebral hyperemia were prevented during HBO2 at 5 and 6 ATA. Tiagabine and the nonselective GAT inhibitor nipecotic acid similarly extended HBO2 seizure latencies. In mice pretreated with tiagabine and exposed to HBO2 at 5 ATA, nuclear and mitochondrial DNA oxidation and astrocytosis was attenuated in the cerebellum and hippocampus. Less oxidant injury in these regions was accompanied by reduced conjugated microtubule-associated protein 1A/1B-light chain 3 (LC3-II), an index of mitophagy, and phosphorylated cAMP response element binding protein (pCREB), an initiator of mitochondrial biogenesis. We conclude that GABA prevents cerebral hyperemia and delays neuroexcitation under extreme HBO2, limiting oxidant damage in the cerebellum and hippocampus, and likely lowering mitophagy flux and initiation of pCREB-initiated mitochondrial biogenesis.

4.
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
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.
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
7.
Obesity (Silver Spring) ; 28(5): 924-931, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32237119

RESUMEN

OBJECTIVE: Carbon monoxide (CO) may counteract obesity and metabolic dysfunction in rodents consuming high-fat diets, but the skeletal effects are not understood. This study investigated whether low-dose inhaled CO (250 ppm) with or without moderate intensity aerobic exercise (3 h/wk) would limit diet-induced obesity and metabolic dysregulation and preserve bone health. METHODS: Obesity-resistant (OR) rats served as controls, and obesity-prone (OP) rats were randomized to sedentary, sedentary plus CO, exercise, or CO plus exercise. For 10 weeks, OP rats consumed a high-fat, high-sucrose diet, whereas OR rats consumed a low-fat control diet. Measurements included indicators of obesity and metabolism, bone turnover markers, femoral geometry and microarchitecture, bone mechanical properties, and tibial morphometry. RESULTS: A high-fat, high-sucrose diet led to obesity, hyperinsulinemia, and hyperleptinemia, without impacting bone. CO alone led only to a modest reduction in weight gain. Exercise attenuated weight gain and improved the metabolic profile; however, bone fragility increased. Combined CO and exercise led to body mass reduction and a metabolic state similar to control OR rats and prevented the exercise-induced increase in bone fragility. CONCLUSIONS: CO and aerobic exercise training prevent obesity and metabolic sequelae of nutrient excess while stabilizing bone physiology.


Asunto(s)
Monóxido de Carbono , Obesidad , Condicionamiento Físico Animal , Animales , Masculino , Ratas , Monóxido de Carbono/farmacología , Monóxido de Carbono/uso terapéutico , Obesidad/prevención & control , Condicionamiento Físico Animal/fisiología
8.
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
9.
Free Radic Biol Med ; 152: 455-461, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31958498

RESUMEN

Mitochondrial oxidant damage, including damage to mitochondrial DNA (mtDNA) is a feature of both severe microbial infections and inflammation arising from sterile (non-infectious) sources such as tissue trauma. Damaged mitochondria release intact or oxidized fragments of mtDNA into the cytoplasm, which represent oxidant injury, and the fragments promote a spontaneous innate immune response, exemplifying a modern frontier of immunological research. MtDNA and mitochondrial-derived oxidants are central factors in activating at least three innate immune pathways involving the TLR9 (Toll-like receptor 9), the NLRP3 (NACHT, LRR and PYD domains-containing protein-3) inflammasome, and the cGAS (cyclic AMP-GMP synthase) pathway. The events that allow mtDNA to escape from damaged mitochondria and from damaged cells are incompletely known, but the presence of cytoplasmic mtDNA and cell-free mtDNA as immune regulators are important for understanding the cell's capacity for protecting mitochondrial quality control (MQC) and cell viability during inflammatory states.


Asunto(s)
ADN Mitocondrial , Oxidantes , ADN Mitocondrial/genética , Inmunidad Innata , Inflamasomas , Mitocondrias/genética
10.
Neurotox Res ; 36(4): 788-795, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31148118

RESUMEN

Hyperbaric oxygen (HBO2) is acutely toxic to the central nervous system, culminating in EEG spikes and tonic-clonic convulsions. GABA enhancers and sodium channel antagonists improve seizure latencies in HBO2 when administered individually, while combining antiepileptic drugs from different functional classes can provide greater seizure latency. We examined the combined effectiveness of GABA enhancers (tiagabine and gabapentin) with sodium channel antagonists (carbamazepine and lamotrigine) in delaying HBO2-induced seizures. A series of experiments in C57BL/6 mice exposed to 100% oxygen at 5 atmospheres absolute (ATA) were performed. We predicted equally effective doses from individual drug-dose response curves, and the combinations of tiagabine + carbamazepine or lamotrigine were tested to determine the maximally effective combined doses to be used in subsequent experiments designed to identify the type of pharmacodynamic interaction for three fixed-ratio combinations (1:3, 1:1, and 3:1) using isobolographic analysis. For both combinations, the maximally effective combined doses increased seizure latency over controls > 5-fold and were determined to interact synergistically for fixed ratios 1:1 and 3:1, additive for 1:3. These results led us to explore whether the benefits of these drug combinations could be extended to the lungs, since a centrally mediated mechanism is believed to mediate hyperoxic-induced cardiogenic lung injury. Indeed, both combinations attenuated bronchoalveolar lavage protein content by ~ 50%. Combining tiagabine with carbamazepine or lamotrigine not only affords greater antiseizure protection in HBO2 but also allows for lower doses to be used, minimizing side effects, and attenuating acute lung injury.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Oxigenoterapia Hiperbárica , Oxígeno/toxicidad , Convulsiones/inducido químicamente , Bloqueadores de los Canales de Sodio/administración & dosificación , Tiagabina/administración & dosificación , Animales , Carbamazepina/administración & dosificación , Gabapentina/administración & dosificación , Lamotrigina/administración & dosificación , Ratones Endogámicos C57BL , Convulsiones/tratamiento farmacológico
11.
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
12.
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
13.
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
14.
JCI Insight ; 3(23)2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30518685

RESUMEN

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a prevalent disease with significant mortality for which no effective pharmacologic therapy exists. Low-dose inhaled carbon monoxide (iCO) confers cytoprotection in preclinical models of sepsis and ARDS. METHODS: We conducted a phase I dose escalation trial to assess feasibility and safety of low-dose iCO administration in patients with sepsis-induced ARDS. Twelve participants were randomized to iCO or placebo air 2:1 in two cohorts. Four subjects each were administered iCO (100 ppm in cohort 1 or 200 ppm in cohort 2) or placebo for 90 minutes for up to 5 consecutive days. Primary outcomes included the incidence of carboxyhemoglobin (COHb) level ≥10%, prespecified administration-associated adverse events (AEs), and severe adverse events (SAEs). Secondary endpoints included the accuracy of the Coburn-Forster-Kane (CFK) equation to predict COHb levels, biomarker levels, and clinical outcomes. RESULTS: No participants exceeded a COHb level of 10%, and there were no administration-associated AEs or study-related SAEs. CO-treated participants had a significant increase in COHb (3.48% ± 0.7% [cohort 1]; 4.9% ± 0.28% [cohort 2]) compared with placebo-treated subjects (1.97% ± 0.39%). The CFK equation was highly accurate at predicting COHb levels, particularly in cohort 2 (R2 = 0.9205; P < 0.0001). Circulating mitochondrial DNA levels were reduced in iCO-treated participants compared with placebo-treated subjects. CONCLUSION: Precise administration of low-dose iCO is feasible, well-tolerated, and appears to be safe in patients with sepsis-induced ARDS. Excellent agreement between predicted and observed COHb should ensure that COHb levels remain in the target range during future efficacy trials. TRIAL REGISTRATION: ClinicalTrials.gov NCT02425579. FUNDING: NIH grants P01HL108801, KL2TR002385, K08HL130557, and K08GM102695.


Asunto(s)
Administración por Inhalación , Monóxido de Carbono/administración & dosificación , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Terapia Respiratoria/métodos , Sepsis/tratamiento farmacológico , Adulto , Anciano , Biomarcadores/sangre , Análisis de los Gases de la Sangre , Carboxihemoglobina , ADN Mitocondrial , Femenino , Humanos , Masculino , Persona de Mediana Edad
15.
J Appl Physiol (1985) ; 125(4): 1296-1304, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30024340

RESUMEN

Exposure to extreme hyperbaric oxygen (HBO2) >5-6 atmospheres absolute (ATA) produces baroreflex impairment, sympathetic hyperactivation, hypertension, tachycardia, and cerebral hyperemia, known as phase II, culminating in seizures. We hypothesized that attenuation of the effects of high sympathetic outflow would preserve regional cerebral blood flow (rCBF) and protect against HBO2-induced seizures. To explore this possibility, we tested four adrenoceptor antagonists in conscious and anesthetized rats exposed to HBO2 at 5 and 6 ATA, respectively: phentolamine (nonselective α1 and α2), prazosin (selective α1), propranolol (nonselective ß1 and ß2), and atenolol (selective ß1). In conscious rats, four drug doses were administered to rats before HBO2 exposures, and seizure latencies were recorded. Drug doses that provided similar protection against seizures were administered before HBO2 exposures in anesthetized rats to determine the effects of adrenoceptor blockade on mean arterial pressure, heart rate, rCBF, and EEG spikes. All four drugs modified cardiovascular and rCBF responses in HBO2 that aligned with epileptiform discharges, but only phentolamine and propranolol effectively increased EEG spike latencies by ~20 and 36 min, respectively. When phentolamine and propranolol were delivered during HBO2 at the onset of phase II, only propranolol led to sustained reductions in heart rate and rCBF, preventing the appearance of epileptiform discharges. The enhanced effectiveness of propranolol may extend beyond ß-adrenoceptor blockade, i.e., membrane stability and reduced metabolic activity. These results indicate that adrenoceptor drug pretreatment will minimize the effects of excessive sympathetic outflow on rCBF and extend HBO2 exposure time.NEW & NOTEWORTHY Blocking adrenergic receptors with phentolamine (nonselective α1 and α2), prazosin (selective α1), propranolol (nonselective ß1 and ß2), and atenolol (selective ß1) modified cardiovascular and regional cerebral blood flow (rCBF) responses in hyperbaric oxygen (HBO2) at 6 atmospheres absolute (ATA); however, only phentolamine and propranolol extended EEG spike latencies. When these two agents were delivered at the onset of sympathetic hyperactivation, only propranolol reduced heart rate and rCBF throughout the exposure and prevented epileptiform discharges. These data validate the strong role of adrenergic control of cardiovascular function and rCBF in extreme HBO2 and the potential use of antiadrenergic drugs to prevent seizures.

16.
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
17.
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
18.
Clin Pharmacol Ther ; 104(3): 553-563, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29238951

RESUMEN

Disruption of microvascular blood flow is a common cause of tissue hypoxia in disease, yet no therapies are available that directly target the microvasculature to improve tissue oxygenation. Red blood cells (RBCs) autoregulate blood flow through S-nitroso-hemoglobin (SNO-Hb)-mediated export of nitric oxide (NO) bioactivity. We therefore tested the idea that pharmacological enhancement of RBCs using the S-nitrosylating agent ethyl nitrite (ENO) may provide a novel approach to improve tissue oxygenation. Serial ENO dosing was carried out in sheep (1-400 ppm) and humans (1-100 ppm) at normoxia and at reduced fraction of inspired oxygen (FiO2 ). ENO increased RBC SNO-Hb levels, corrected hypoxia-induced deficits in tissue oxygenation, and improved measures of oxygen utilization in both species. No adverse effects or safety concerns were identified. Inasmuch as impaired oxygenation is a major cause of morbidity and mortality, ENO may have widespread therapeutic utility, providing a first-in-class agent targeting the microvasculature.


Asunto(s)
Eritrocitos/efectos de los fármacos , Hipoxia/tratamiento farmacológico , Nitritos/administración & dosificación , Oxígeno/sangre , Vasodilatación/efectos de los fármacos , Vasodilatadores/administración & dosificación , Adolescente , Adulto , Animales , Biomarcadores/sangre , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Femenino , Hemoglobinas/metabolismo , Humanos , Hipoxia/sangre , Hipoxia/fisiopatología , Masculino , Óxido Nítrico/sangre , Nitritos/efectos adversos , Oveja Doméstica , Factores de Tiempo , Vasodilatadores/efectos adversos , Adulto Joven
19.
Clin Pharmacol Ther ; 103(5): 888-898, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28857147

RESUMEN

We hypothesized that concomitant pharmacological inhibition of the endothelin and adenosine pathway is safe and improves exercise performance in hypoxic humans, via a mechanism that does not involve augmentation of blood oxygenation. To test this hypothesis, we established safety and drug interactions for aminophylline (500 mg) plus ambrisentan (5 mg) in normoxic volunteers. Subsequently, a placebo-controlled study was employed to test the combination in healthy resting and exercising volunteers at simulated altitude (4,267 m). No serious adverse events occurred. Drug interaction was minimal or absent. Aminophylline alleviated hypoxia-induced headaches. Aminophylline, ambrisentan, and their combination all significantly (P < 0.05 vs. placebo) improved submaximal hypoxic exercise performance (19.5, 20.6, and 19.1% >placebo). Single-dose ambrisentan increased blood oxygenation in resting, hypoxic subjects. We conclude that combined aminophylline and ambrisentan offer promise to safely increase exercise capacity in hypoxemic humans without relying on increasing blood oxygen availability.


Asunto(s)
Aminofilina/efectos adversos , Aminofilina/uso terapéutico , Endotelinas/efectos de los fármacos , Ejercicio Físico/fisiología , Hipoxia/tratamiento farmacológico , Fenilpropionatos/efectos adversos , Fenilpropionatos/uso terapéutico , Piridazinas/efectos adversos , Piridazinas/uso terapéutico , Adenosina/metabolismo , Adolescente , Adulto , Altitud , Método Doble Ciego , Quimioterapia Combinada/efectos adversos , Endotelinas/metabolismo , Femenino , Humanos , Hipoxia/metabolismo , Masculino , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Adulto Joven
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...