Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Bases de datos
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Am J Physiol Lung Cell Mol Physiol ; 309(8): L857-71, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26186946

RESUMEN

The heme oxygenase-1 (HO-1)/carbon monoxide (CO) system induces mitochondrial biogenesis, but its biological impact in human skeletal muscle is uncertain. The enzyme system generates CO, which stimulates mitochondrial proliferation in normal muscle. Here we examined whether CO breathing can be used to produce a coordinated metabolic and vascular response in human skeletal muscle. In 19 healthy subjects, we performed vastus lateralis muscle biopsies and tested one-legged maximal O2 uptake (V̇o2max) before and after breathing air or CO (200 ppm) for 1 h daily for 5 days. In response to CO, there was robust HO-1 induction along with increased mRNA levels for nuclear-encoded mitochondrial transcription factor A (Tfam), cytochrome c, cytochrome oxidase subunit IV (COX IV), and mitochondrial-encoded COX I and NADH dehydrogenase subunit 1 (NDI). CO breathing did not increase V̇o2max (1.96 ± 0.51 pre-CO, 1.87 ± 0.50 post-CO l/min; P = not significant) but did increase muscle citrate synthase, mitochondrial density (139.0 ± 34.9 pre-CO, 219.0 ± 36.2 post-CO; no. of mitochondrial profiles/field), myoglobin content and glucose transporter (GLUT4) protein level and led to GLUT4 localization to the myocyte membrane, all consistent with expansion of the tissue O2 transport system. These responses were attended by increased cluster of differentiation 31 (CD31)-positive muscle capillaries (1.78 ± 0.16 pre-CO, 2.37 ± 0.59 post-CO; capillaries/muscle fiber), implying the enrichment of microvascular O2 reserve. The findings support that induction of the HO-1/CO system by CO not only improves muscle mitochondrial density, but regulates myoglobin content, GLUT4 localization, and capillarity in accordance with current concepts of skeletal muscle plasticity.


Asunto(s)
Monóxido de Carbono/metabolismo , Hemo-Oxigenasa 1/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Adolescente , Adulto , Capilares/anatomía & histología , ADN Mitocondrial/genética , Prueba de Esfuerzo , Femenino , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Masculino , Microscopía Electrónica de Transmisión , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/ultraestructura , Proteínas Musculares/metabolismo , Músculo Esquelético/ultraestructura , Consumo de Oxígeno , Músculo Cuádriceps/irrigación sanguínea , Músculo Cuádriceps/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adulto Joven
12.
Am J Physiol Lung Cell Mol Physiol ; 309(8): L834-46, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26320156

RESUMEN

Inhaled carbon monoxide (CO) gas has therapeutic potential for patients with acute respiratory distress syndrome if a safe, evidence-based dosing strategy and a ventilator-compatible CO delivery system can be developed. In this study, we used a clinically relevant baboon model of Streptococcus pneumoniae pneumonia to 1) test a novel, ventilator-compatible CO delivery system; 2) establish a safe and effective CO dosing regimen; and 3) investigate the local and systemic effects of CO therapy on inflammation and acute lung injury (ALI). Animals were inoculated with S. pneumoniae (10(8)-10(9) CFU) (n = 14) or saline vehicle (n = 5); in a subset with pneumonia (n = 5), we administered low-dose, inhaled CO gas (100-300 ppm × 60-90 min) at 0, 6, 24, and/or 48 h postinoculation and serially measured blood carboxyhemoglobin (COHb) levels. We found that CO inhalation at 200 ppm for 60 min is well tolerated and achieves a COHb of 6-8% with ambient CO levels ≤ 1 ppm. The COHb level measured at 20 min predicted the 60-min COHb level by the Coburn-Forster-Kane equation with high accuracy. Animals given inhaled CO + antibiotics displayed significantly less ALI at 8 days postinoculation compared with antibiotics alone. Inhaled CO was associated with activation of mitochondrial biogenesis in the lung and with augmentation of renal antioxidative programs. These data support the feasibility of safely delivering inhaled CO gas during mechanical ventilation and provide preliminary evidence that CO may accelerate the resolution of ALI in a clinically relevant nonhuman primate pneumonia model.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/terapia , Monóxido de Carbono/administración & dosificación , Neumonía Neumocócica/complicaciones , Neumonía Neumocócica/terapia , Lesión Pulmonar Aguda/sangre , Administración por Inhalación , Animales , Antibacterianos/administración & dosificación , Antioxidantes/metabolismo , Carboxihemoglobina/metabolismo , Modelos Animales de Enfermedad , Diseño de Equipo , Humanos , Riñón/metabolismo , Pulmón/patología , Masculino , Papio , Neumonía Neumocócica/sangre , Respiración Artificial , Terapia Respiratoria/instrumentación , Sepsis/etiología , Sepsis/metabolismo , Sepsis/terapia
13.
Biochim Biophys Acta ; 1820(4): 532-41, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22265687

RESUMEN

BACKGROUND: Cells avoid major mitochondrial damage and energy failure during systemic inflammatory states, such as severe acute infections, by specific targeting of the inflammatory response and by inducing anti-inflammatory and anti-oxidant defenses. Recent evidence indicates that these cell defenses also include mitochondrial biogenesis and the clearance of damaged mitochondria through autophagy. SCOPE OF REVIEW: This review addresses a group of transcriptional signaling mechanisms that engage mitochondrial biogenesis, including energy-sensing and redox-regulated transcription factors and co-activators, after major inflammatory events. MAJOR CONCLUSIONS: Stimulation of the innate immune system by activation of toll-like receptors (TLR) generates pro-inflammatory mediators, such as tumor necrosis factor-α (TNF-α)and interleukin-1ß (IL-1ß), necessary for optimal host defense, but which also contribute to mitochondrial damage through oxidative stress and other mechanisms. To protect its energy supply, host cells sense mitochondrial damage and initiate mitochondrial biogenesis under the control of an inducible transcriptional program that also activates anti-oxidant and anti-inflammatory gene expression. This multifunctional network not only increases cellular resistance to metabolic failure, oxidative stress, and cell death, but promotes immune tolerance as shown in the graphical abstract. GENERAL SIGNIFICANCE: The post-inflammatory induction of mitochondrial biogenesis supports metabolic function and cell viability while helping to control inflammation. In clinical settings, patients recovering from severe systemic infections may develop transient immune suppression, placing them at risk for recurrent infection, but there may be therapeutic opportunities to enhance mitochondrial quality control that would improve the resolution of life-threatening host responses to such infections.


Asunto(s)
Inflamación/metabolismo , Mitocondrias/metabolismo , Transcripción Genética , Animales , Calcio/metabolismo , Supervivencia Celular , Regulación de la Expresión Génica , Inmunidad Innata , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación/inmunología , Mediadores de Inflamación , Interleucina-1beta/biosíntesis , Ratones , Mitocondrias/genética , Estrés Oxidativo , Transducción de Señal , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
14.
Gastroenterology ; 143(5): 1319-1329.e11, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22885334

RESUMEN

BACKGROUND & AIMS: The pathogenesis of cirrhosis, a disabling outcome of defective liver repair, involves deregulated accumulation of myofibroblasts derived from quiescent hepatic stellate cells (HSCs), but the mechanisms that control transdifferentiation of HSCs are poorly understood. We investigated whether the Hedgehog (Hh) pathway controls the fate of HSCs by regulating metabolism. METHODS: Microarray, quantitative polymerase chain reaction, and immunoblot analyses were used to identify metabolic genes that were differentially expressed in quiescent vs myofibroblast HSCs. Glycolysis and lactate production were disrupted in HSCs to determine if metabolism influenced transdifferentiation. Hh signaling and hypoxia-inducible factor 1α (HIF1α) activity were altered to identify factors that alter glycolytic activity. Changes in expression of genes that regulate glycolysis were quantified and localized in biopsy samples from patients with cirrhosis and liver samples from mice following administration of CCl(4) or bile duct ligation. Mice were given systemic inhibitors of Hh to determine if they affect glycolytic activity of the hepatic stroma; Hh signaling was also conditionally disrupted in myofibroblasts to determine the effects of glycolytic activity. RESULTS: Transdifferentiation of cultured, quiescent HSCs into myofibroblasts induced glycolysis and caused lactate accumulation. Increased expression of genes that regulate glycolysis required Hh signaling and involved induction of HIF1α. Inhibitors of Hh signaling, HIF1α, glycolysis, or lactate accumulation converted myofibroblasts to quiescent HSCs. In diseased livers of animals and patients, numbers of glycolytic stromal cells were associated with the severity of fibrosis. Conditional disruption of Hh signaling in myofibroblasts reduced numbers of glycolytic myofibroblasts and liver fibrosis in mice; similar effects were observed following administration of pharmacologic inhibitors of Hh. CONCLUSIONS: Hedgehog signaling controls the fate of HSCs by regulating metabolism. These findings might be applied to diagnosis and treatment of patients with cirrhosis.


Asunto(s)
Transdiferenciación Celular/genética , Regulación de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Células Estrelladas Hepáticas/metabolismo , Miofibroblastos/metabolismo , Transducción de Señal/genética , Actinas/genética , Actinas/metabolismo , Animales , Conductos Biliares , Tetracloruro de Carbono , Células Cultivadas , Perfilación de la Expresión Génica , Glucólisis/genética , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/enzimología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ácido Láctico/metabolismo , Ligadura , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias , Miofibroblastos/enzimología , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , ARN Mensajero/metabolismo , Ratas , Factores de Tiempo
15.
Am J Respir Crit Care Med ; 185(8): 851-61, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22312014

RESUMEN

RATIONALE: Mitochondrial damage is an important component of multiple organ failure syndrome, a highly lethal complication of severe sepsis that lacks specific therapy. Mitochondrial quality control is regulated in part by the heme oxygenase-1 (HO-1; Hmox1) system through the redox-regulated NF-E2-related factor-2 (Nrf2) transcription factor, but its role in mitochondrial biogenesis in Staphylococcus aureus sepsis is unknown. OBJECTIVES: To test the hypothesis that Nrf2-dependent up-regulation of the HO-1/carbon monoxide (CO) system would preserve mitochondrial biogenesis and rescue mice from lethal S. aureus sepsis. METHODS: A controlled murine S. aureus peritonitis model with and without inhaled CO was examined for HO-1 and Nrf2 regulation of mitochondrial biogenesis and the resolution of hepatic mitochondrial damage. MEASUREMENTS AND MAIN RESULTS: Sepsis survival was significantly enhanced using inhaled CO (250 ppm once-daily for 1 h), and linked mechanistically to Hmox1 induction and mitochondrial HO activity through Nrf2 transcriptional and Akt kinase activity. HO-1/CO stimulated Nrf2-dependent gene expression and nuclear accumulation of nuclear respiratory factor-1, -2α (Gabpa), and peroxisome proliferator-activated receptor gamma coactivator-1α; increased mitochondrial transcription factor-A and citrate synthase protein levels; and augmented mtDNA copy number. CO enhanced antiinflammatory IL-10 and reduced proinflammatory tumor necrosis factor-α production. By contrast, Nrf2(-/-) and Akt1(-/-) mice lacked CO induction of Hmox1 and mitochondrial biogenesis, and CO rescued neither strain from S. aureus sepsis. CONCLUSIONS: We identify an inducible Nrf2/HO-1 regulatory cycle for mitochondrial biogenesis that is prosurvival and counter-inflammatory in sepsis, and describe targeted induction of mitochondrial biogenesis as a potential multiple organ failure therapy.


Asunto(s)
Monóxido de Carbono/farmacología , Hemo-Oxigenasa 1/metabolismo , Mitocondrias/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Sepsis/enzimología , Infecciones Estafilocócicas/terapia , Administración por Inhalación , Animales , Western Blotting , Monóxido de Carbono/metabolismo , Modelos Animales de Enfermedad , Femenino , Hemo-Oxigenasa 1/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Factor 2 Relacionado con NF-E2/genética , Biogénesis de Organelos , Peritonitis/tratamiento farmacológico , Peritonitis/microbiología , Peritonitis/mortalidad , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Medición de Riesgo , Sepsis/genética , Sepsis/mortalidad , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Tasa de Supervivencia , Regulación hacia Arriba
16.
J Biol Chem ; 286(18): 16374-85, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21454555

RESUMEN

The induction of heme oxygenase-1 (HO-1; Hmox1) by inflammation, for instance in sepsis, is associated both with an anti-inflammatory response and with mitochondrial biogenesis. Here, we tested the idea that HO-1, acting through the Nfe2l2 (Nrf2) transcription factor, links anti-inflammatory cytokine expression to activation of mitochondrial biogenesis. HO-1 induction after LPS stimulated anti-inflammatory IL-10 and IL-1 receptor antagonist (IL-1Ra) expression in mouse liver, human HepG2 cells, and mouse J774.1 macrophages but blunted tumor necrosis factor-α expression. This was accompanied by nuclear Nfe2l2 accumulation and led us to identify abundant Nfe2l2 and other mitochondrial biogenesis transcription factor binding sites in the promoter regions of IL10 and IL1Ra compared with pro-inflammatory genes regulated by NF-κΒ. Mechanistically, HO-1, through its CO product, enabled these transcription factors to bind the core IL10 and IL1Ra promoters, which for IL10 included Nfe2l2, nuclear respiratory factor (NRF)-2 (Gabpa), and MEF2, and for IL1Ra, included NRF-1 and MEF2. In cells, Hmox1 or Nfe2l2 RNA silencing prevented IL-10 and IL-1Ra up-regulation, and HO-1 induction failed post-LPS in Nfe2l2-silenced cells and post-sepsis in Nfe2l2(-/-) mice. Nfe2l2(-/-) mice compared with WT mice, showed more liver damage, higher mortality, and ineffective CO rescue in sepsis. Nfe2l2(-/-) mice in sepsis also generated higher hepatic TNF-α mRNA levels, lower NRF-1 and PGC-1α mRNA levels, and no enhancement of anti-inflammatory Il10, Socs3, or bcl-x(L) gene expression. These findings disclose a highly structured transcriptional network that couples mitochondrial biogenesis to counter-inflammation with major implications for immune suppression in sepsis.


Asunto(s)
Citocinas/biosíntesis , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias Hepáticas/enzimología , Sepsis/enzimología , Factores de Transcripción/metabolismo , Animales , Monóxido de Carbono/metabolismo , Citocinas/genética , Hemo-Oxigenasa 1/genética , Células Hep G2 , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Sepsis/genética , Factores de Transcripción/genética
17.
J Cell Sci ; 123(Pt 15): 2565-75, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20587593

RESUMEN

The nuclear respiratory factor-1 (NRF1) gene is activated by lipopolysaccharide (LPS), which might reflect TLR4-mediated mitigation of cellular inflammatory damage via initiation of mitochondrial biogenesis. To test this hypothesis, we examined NRF1 promoter regulation by NFκB, and identified interspecies-conserved κB-responsive promoter and intronic elements in the NRF1 locus. In mice, activation of Nrf1 and its downstream target, Tfam, by Escherichia coli was contingent on NFκB, and in LPS-treated hepatocytes, NFκB served as an NRF1 enhancer element in conjunction with NFκB promoter binding. Unexpectedly, optimal NRF1 promoter activity after LPS also required binding by the energy-state-dependent transcription factor CREB. EMSA and ChIP assays confirmed p65 and CREB binding to the NRF1 promoter and p65 binding to intron 1. Functionality for both transcription factors was validated by gene-knockdown studies. LPS regulation of NRF1 led to mtDNA-encoded gene expression and expansion of mtDNA copy number. In cells expressing plasmid constructs containing the NRF-1 promoter and GFP, LPS-dependent reporter activity was abolished by cis-acting κB-element mutations, and nuclear accumulation of NFκB and CREB demonstrated dependence on mitochondrial H(2)O(2). These findings indicate that TLR4-dependent NFκB and CREB activation co-regulate the NRF1 promoter with NFκB intronic enhancement and redox-regulated nuclear translocation, leading to downstream target-gene expression, and identify NRF-1 as an early-phase component of the host antibacterial defenses.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Factor Nuclear 1 de Respiración/metabolismo , Animales , Inmunoprecipitación de Cromatina , Biología Computacional , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , ADN Mitocondrial/genética , Ensayo de Cambio de Movilidad Electroforética , Células Hep G2 , Humanos , Immunoblotting , Inflamación/inducido químicamente , Intrones/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Factor Nuclear 1 de Respiración/genética , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Unión Proteica/fisiología , Sulfonas/farmacología
18.
Circ Res ; 106(11): 1722-30, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20395592

RESUMEN

RATIONALE: Erythropoietin (EPO) is often administered to cardiac patients with anemia, particularly from chronic kidney disease, and stimulation of erythropoiesis may stabilize left ventricular and renal function by recruiting protective effects beyond the correction of anemia. OBJECTIVE: We examined the hypothesis that EPO receptor (EpoR) ligand-binding, which activates endothelial NO synthase (eNOS), regulates the prosurvival program of mitochondrial biogenesis in the heart. METHODS AND RESULTS: We investigated the effects of EPO on mitochondrial biogenesis over 14 days in healthy mice. Mice expressing a mitochondrial green fluorescent protein reporter construct demonstrated sharp increases in myocardial mitochondrial density after 3 days of EPO administration that peaked at 7 days and surpassed hepatic or renal effects and anteceded significant increases in blood hemoglobin content. Quantitatively, in wild-type mice, complex II activity, state 3 respiration, and mtDNA copy number increased significantly; also, resting energy expenditure and natural running speed improved, with no evidence of an increase in left ventricular mass index. Mechanistically, EPO activated cardiac mitochondrial biogenesis by enhancement of nuclear respiratory factor-1, PGC-1alpha (peroxisome proliferator-activated receptor gamma coactivator 1alpha), and mitochondrial transcription factor-A gene expression in wild-type but not in eNOS(-/-) or protein kinase B (Akt1)(-/-) mice. EpoR was required, because EpoR silencing in cardiomyocytes blocked EPO-mediated nuclear translocation of nuclear respiratory factor-1. CONCLUSIONS: These findings support a new physiological and protective role for EPO, acting through its cell surface receptor and eNOS-Akt1 signal transduction, in matching cardiac mitochondrial mass to the convective O(2) transport capacity as erythrocyte mass expands.


Asunto(s)
Eritrocitos/efectos de los fármacos , Eritropoyetina/farmacología , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Ecocardiografía , Complejo II de Transporte de Electrones/metabolismo , Metabolismo Energético/efectos de los fármacos , Eritrocitos/metabolismo , Eritropoyetina/administración & dosificación , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Hemoglobinas/metabolismo , Humanos , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Factor Nuclear 1 de Respiración/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Interferencia de ARN , Receptores de Eritropoyetina/efectos de los fármacos , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
19.
Am J Respir Crit Care Med ; 183(2): 226-33, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20732986

RESUMEN

RATIONALE: Damage to mitochondrial DNA (mtDNA) by the production of reactive oxygen species during inflammatory states, such as sepsis, is repaired by poorly understood mechanisms. OBJECTIVES: To test the hypothesis that the DNA repair enzyme, 8-oxoguanine DNA glycosylase (OGG1), contributes to mtDNA repair in sepsis. METHODS: Using a well-characterized mouse model of Staphylococcus aureus sepsis, we analyzed molecular markers for mitochondrial biogenesis and OGG1 translocation into liver mitochondria as well as OGG1 mRNA expression at 0, 24, 48, and 72 hours after infection. The effects of OGG1 RNA silencing on mtDNA content were determined in control, tumor necrosis factor-α, and peptidoglycan-exposed rat hepatoma cells. Based on in situ analysis of the OGG1 promoter region, chromatin immunoprecipitation assays were performed for nuclear respiratory factor (NRF)-1 and NRF-2α GA-binding protein (GABP) binding to the promoter of OGG1. MEASUREMENTS AND MAIN RESULTS: Mice infected with 10(7) cfu S. aureus intraperitoneally demonstrated hepatic oxidative mtDNA damage and significantly lower hepatic mtDNA content as well as increased mitochondrial OGG1 protein and enzyme activity compared with control mice. The infection also caused increases in hepatic OGG1 transcript levels and NRF-1 and NRF-2α transcript and protein levels. A bioinformatics analysis of the Ogg1 gene locus identified several promoter sites containing NRF-1 and NRF-2α DNA binding motifs, and chromatin immunoprecipitation assays confirmed in situ binding of both transcription factors to the Ogg1 promoter within 24 hours of infection. CONCLUSIONS: These studies identify OGG1 as an early mitochondrial response protein during sepsis under regulation by the NRF-1 and NRF-2α transcription factors that regulate mitochondrial biogenesis.


Asunto(s)
ADN Glicosilasas/metabolismo , Reparación del ADN , ADN Mitocondrial/metabolismo , Mitocondrias Hepáticas/enzimología , Sepsis/enzimología , Staphylococcus aureus , Animales , Western Blotting , Modelos Animales de Enfermedad , Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Inmunoprecipitación/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Factor Nuclear 1 de Respiración/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sepsis/microbiología
20.
J Oral Maxillofac Surg ; 70(7): 1573-83, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22698292

RESUMEN

PURPOSE: This study tested hyperbaric oxygen (HBO) as an adjunct to surgery and antibiotics in the treatment of bisphosphonate-related osteonecrosis of the jaw (ONJ) and evaluated its effects on gingival healing, pain, and quality of life. MATERIALS AND METHODS: The investigators implemented a randomized controlled trial and enrolled a sample composed of patients with ONJ, where the predictor variable was HBO administered at 2 atm twice a day for 40 treatments as an adjunct to conventional therapy of surgery and antibiotics versus conventional therapy alone. Over the next 24 months, oral lesion size and number, pain, and quality of life were assessed. RESULTS: Forty-six patients (mean age, 66 yrs; 57% women) contributed data to the trial. There were no statistically significant differences in the distribution of variables used to assess randomization success between the HBO and standard treatment groups. Seventeen of 25 HBO-treated patients (68%) improved versus 8 of 21 controls (38.1%; P = .043, χ(2) test). Mean time to improvement was 39.7 weeks (95% confidence interval [CI], 22.4 to 57.0 weeks) for HBO-treated patients versus 67.9 weeks (95 CI, 48.4 to 87.5 weeks) for controls (P = .03, log-rank test). However, complete gingival healing occurred in only 14 of 25 HBO-treated patients (52%) versus 7 of 21 controls (33.3%; P = .203, χ(2) test), and time to healing was 59 weeks (95% CI, 42.8% to 75.8%) for HBO-treated patients versus 70 weeks (95 CI, 52.2% to 88.36%) for controls (P = .32, log-rank test). Pain decreased faster for HBO-treated subjects (P < .01, linear regression). Quality-of-life scores for physical health (P = .002) and perceived health (P = .043) decreased at 6 months for control group but for not the HBO group. CONCLUSIONS: ONJ is multifactorial and no single treatment modality is likely to reverse it; however, it is treatable and even advanced presentations can improve with intensive multimodal therapy. Clinically, HBO appears to be a useful adjunct to ONJ treatment, particularly for more severe cases, although this study was underpowered to fully support this claim.


Asunto(s)
Antibacterianos/uso terapéutico , Osteonecrosis de los Maxilares Asociada a Difosfonatos/terapia , Desbridamiento/métodos , Oxigenoterapia Hiperbárica , Anciano , Alendronato/efectos adversos , Osteonecrosis de los Maxilares Asociada a Difosfonatos/tratamiento farmacológico , Osteonecrosis de los Maxilares Asociada a Difosfonatos/cirugía , Conservadores de la Densidad Ósea/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Terapia Combinada , Difosfonatos/efectos adversos , Femenino , Estudios de Seguimiento , Encía/patología , Humanos , Imidazoles/efectos adversos , Masculino , Mieloma Múltiple/tratamiento farmacológico , Osteoporosis/tratamiento farmacológico , Manejo del Dolor , Pamidronato , Estudios Prospectivos , Calidad de Vida , Resultado del Tratamiento , Cicatrización de Heridas/fisiología , Ácido Zoledrónico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA