Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
J Immunol ; 204(6): 1474-1485, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31996456

RESUMO

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.


Assuntos
Hipertensão Pulmonar/imunologia , Hipóxia/complicações , Macrófagos Alveolares/imunologia , Monócitos/imunologia , Remodelação Vascular/imunologia , Animais , Antígenos Ly/metabolismo , Transplante de Medula Óssea , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Humanos , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/cirurgia , Hipóxia/imunologia , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/irrigação sanguínea , Pulmão/imunologia , Pulmão/patologia , Transplante de Pulmão , Macrófagos Alveolares/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/imunologia , Artéria Pulmonar/patologia , Quimeras de Transplante/imunologia , Remodelação Vascular/genética
2.
Crit Care Med ; 47(5): 651-658, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30730439

RESUMO

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.


Assuntos
DNA Mitocondrial/sangue , Leucócitos Mononucleares/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Sepse/metabolismo , Adulto , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mitocôndrias/genética , Doenças Mitocondriais/sangue , Doenças Mitocondriais/genética , Estudos Prospectivos , Reação em Cadeia da Polimerase em Tempo Real , Sepse/sangue , Sepse/genética
3.
Proc Natl Acad Sci U S A ; 116(5): 1603-1612, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30655340

RESUMO

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.


Assuntos
Pulmão/metabolismo , Pulmão/fisiopatologia , Pneumonia Bacteriana/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Regeneração/fisiologia , Células-Tronco/metabolismo , Uteroglobina/metabolismo , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/fisiologia , Animais , Bronquíolos/metabolismo , Bronquíolos/fisiopatologia , Diferenciação Celular/fisiologia , Linhagem Celular , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Bacteriana/fisiopatologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/fisiopatologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/fisiopatologia , Células-Tronco/fisiologia
4.
Nitric Oxide ; 74: 56-64, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29355776

RESUMO

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.


Assuntos
Contração Muscular , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , S-Nitrosotióis/metabolismo , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Células HEK293 , Humanos , Ratos
5.
Exp Cell Res ; 363(2): 151-159, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29291399

RESUMO

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.


Assuntos
Morte Celular/efeitos dos fármacos , Micropartículas Derivadas de Células/efeitos dos fármacos , Micropartículas Derivadas de Células/metabolismo , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Animais , Apoptose/efeitos dos fármacos , Inibidores de Caspase/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Necrose/metabolismo , Células RAW 264.7/metabolismo , Estaurosporina/farmacologia
6.
Clin Pharmacol Ther ; 103(5): 888-898, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28857147

RESUMO

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.


Assuntos
Aminofilina/efeitos adversos , Aminofilina/uso terapêutico , Endotelinas/efeitos dos fármacos , Exercício Físico/fisiologia , Hipóxia/tratamento farmacológico , Fenilpropionatos/efeitos adversos , Fenilpropionatos/uso terapêutico , Piridazinas/efeitos adversos , Piridazinas/uso terapêutico , Adenosina/metabolismo , Adolescente , Adulto , Altitude , Método Duplo-Cego , Quimioterapia Combinada/efeitos adversos , Endotelinas/metabolismo , Feminino , Humanos , Hipóxia/metabolismo , Masculino , Pessoa de Meia-Idade , Transdução de Sinais/efeitos dos fármacos , Adulto Jovem
7.
Am J Respir Cell Mol Biol ; 56(3): 300-309, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27636016

RESUMO

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


Assuntos
Proteínas Relacionadas à Autofagia/metabolismo , Autofagia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Mitocôndrias/metabolismo , Estresse Oxidativo , Trifosfato de Adenosina/metabolismo , Animais , Antioxidantes/metabolismo , Autofagia/efeitos dos fármacos , Monóxido de Carbono/farmacologia , Células Cultivadas , Espaço Intracelular/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fenótipo
8.
J Clin Invest ; 126(12): 4402-4403, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27841761

RESUMO

Nitric oxide (NO) is a potent mediator of blood vessel dilation and is released by several cell sources. Red blood cells (rbc) release NO when hemoglobin that has been S-nitrosylated at Cys93 of the ß-chain (ßCys93) transitions from the oxygenated form to the deoxygenated form. This transition occurs in response to reduced tissue oxygenation and is an important physiologic regulator of hypoxic vasodilation. In this issue of the JCI, Zhang and colleagues demonstrate that S-nitrosylation of hemoglobin at ßCys93 is important for tissue oxygenation after cardiac injury. Mice harboring mutations that prevent S-nitrosylation of ßCys93 had higher rates of morbidity and mortality following cardiac injury compared with WT; however, adaptive cardiac vascularization was increased in some mutant mice and reduced cardiac injury in these animals. The results of this study reveal a previously unexplored role of S-nitrosylated hemoglobin in cardioprotection.


Assuntos
Eritrócitos/metabolismo , Hemoglobinas/metabolismo , Hipóxia/metabolismo , Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Vasodilatação , Substituição de Aminoácidos , Animais , Cardiotônicos/metabolismo , Cisteína/genética , Cisteína/metabolismo , Hemoglobinas/genética , Humanos , Hipóxia/genética , Hipóxia/patologia , Camundongos , Camundongos Mutantes , Mutação de Sentido Incorreto , Miocárdio/patologia , Oxirredução
9.
Pharmacol Rev ; 68(1): 20-48, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26589414

RESUMO

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.


Assuntos
Mitocôndrias/metabolismo , Doenças Mitocondriais/fisiopatologia , Monóxido de Carbono/metabolismo , Eritropoetina/metabolismo , Estrogênios/metabolismo , Sequestradores de Radicais Livres/metabolismo , Heme Oxigenase-1/metabolismo , Humanos , Sulfeto de Hidrogênio/metabolismo , Inflamassomos/metabolismo , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Doenças Mitocondriais/induzido quimicamente , Mitofagia/fisiologia , Óxido Nítrico/metabolismo , Fosforilação Oxidativa , Polifenóis/metabolismo , Hormônios Tireóideos/metabolismo
10.
Am J Respir Crit Care Med ; 193(5): 552-60, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26488115

RESUMO

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.


Assuntos
Brônquios/metabolismo , Hipóxia Celular/genética , Pneumopatias/cirurgia , Transplante de Pulmão , Complicações Pós-Operatórias/genética , Insuficiência Respiratória/genética , Transplantes/metabolismo , Adulto , Idoso , Brônquios/irrigação sanguínea , Brônquios/patologia , Constrição Patológica/genética , Fibrose Cística/cirurgia , Feminino , Expressão Gênica , Heme Oxigenase-1/genética , Humanos , Fibrose Pulmonar Idiopática/cirurgia , Tempo de Internação , Doenças Pulmonares Intersticiais/cirurgia , Masculino , Pessoa de Meia-Idade , Necrose/genética , Oximetria , Prognóstico , Doença Pulmonar Obstrutiva Crônica/cirurgia , Reação em Cadeia da Polimerase em Tempo Real , Receptor TIE-2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoidose Pulmonar/cirurgia , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
11.
Am J Physiol Lung Cell Mol Physiol ; 307(10): L746-57, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25217660

RESUMO

Polymorphonuclear leukocyte (PMN)-mediated acute lung injury from ischemia/reperfusion (I/R) remains a major cause of morbidity and mortality in critical care medicine. Here, we report that inhaled low-dose carbon monoxide (CO) and intravenous resolvin D1 (RvD1) in mice each reduced PMN-mediated acute lung injury from I/R. Inhaled CO (125-250 ppm) and RvD1 (250-500 ng) each reduced PMN lung infiltration and gave additive lung protection. In mouse whole blood, CO and RvD1 attenuated PMN-platelet aggregates, reducing leukotrienes (LTs) and thromboxane B2 (TxB2) in I/R lungs. With human whole blood, CO (125-250 ppm) decreased PMN-platelet aggregates, expression of adhesion molecules, and cysteinyl LTs, as well as TxB2. RvD1 (1-100 nM) also dose dependently reduced platelet activating factor-stimulated PMN-platelet aggregates in human whole blood. In nonhuman primate (baboon) lung infection with Streptococcus pneumoniae, inhaled CO reduced urinary cysteinyl LTs. These results demonstrate lung protection by low-dose inhaled CO as well as RvD1 that each reduced PMN-mediated acute tissue injury, PMN-platelet interactions, and production of both cysteinyl LTs and TxB2. Together they suggest a potential therapeutic role of low-dose inhaled CO in organ protection, as demonstrated using mouse I/R-initiated lung injury, baboon infections, and human whole blood.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Antimetabólitos/farmacologia , Monóxido de Carbono/farmacologia , Comunicação Celular/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Leucotrienos/metabolismo , Pulmão/metabolismo , Tromboxano B2/metabolismo , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Pulmão/patologia , Masculino , Camundongos , Papio , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/patologia , Streptococcus pneumoniae/metabolismo
12.
Am J Respir Cell Mol Biol ; 48(5): 647-54, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23492191

RESUMO

In pulmonary arterial hypertension (PAH), there is overexpression of the chemokine, C-C chemokine ligand type 2 (CCL2), and infiltration of myeloid cells into the pulmonary vasculature. Inhibition of CCL2 in animals decreases PAH, suggesting that the CCL2 receptor (CCR2) plays a role in PAH development. To test this hypothesis, we exposed wild-type (WT) and CCR2-deficient (Ccr2(-/-)) mice to chronic hypobaric hypoxia to induce PAH. After hypoxic stress, Ccr2(-/-) mice displayed a more severe PAH phenotype, as demonstrated by increased right ventricular (RV) systolic pressures, RV hypertrophy, and tachycardia relative to WT mice. However, these mice also exhibited increased RV systolic pressures and increased pulmonary artery muscularization under normoxic conditions. Moreover, Ccr2(-/-) mice displayed decreased pulmonary vascular branching at 3 weeks of age and increased vascular muscularization at birth, suggesting that an abnormality in pulmonary vascular development leads to spontaneous PAH in these animals. No significant differences in cytokine responses were observed between WT and Ccr2(-/-) mice during either normoxia or hypoxia. However, Ccr2(-/-) mice displayed increased Notch-3 signaling and dysregulated Notch ligand expression, suggesting a possible cause for their abnormal pulmonary vascular development. Our findings imply that CCR2 does not directly contribute to the development of PAH, but does play a previously unrecognized role in pulmonary vasculature development and remodeling wherein the absence of CCR2 results in spontaneous PAH, most likely via dysregulation of Notch signaling. Our results demonstrate that CCR2 has impacts beyond leukocyte recruitment, and is required for the proper expression of Notch signaling molecules.


Assuntos
Hipertensão Pulmonar/metabolismo , Receptores CCR2/deficiência , Receptores Notch/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Animais , Arteríolas/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Células Dendríticas/imunologia , Hipertensão Pulmonar Primária Familiar , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/imunologia , Hipóxia/complicações , Hipóxia/imunologia , Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Jagged-2 , Pulmão/irrigação sanguínea , Macrófagos/imunologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Receptor Notch3 , Receptores CCR2/genética , Proteínas Serrate-Jagged
13.
Undersea Hyperb Med ; 39(5): 873-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23045915

RESUMO

INTRODUCTION: Supplemental oxygen has been reported to cause pulmonary complications after bleomycin. We describe the safe administration of hyperbaric oxygen (HBO2) after bleomycin in 15 patients. METHODS: Paper and electronic records were reviewed for bleomycin-exposed patients at the Duke Center for Hyperbaric Medicine and Environmental Physiology from 1979 to 2010. RESULTS: Fourteen bleomycin-exposed patients received HBO2 at Duke under a special-precautions protocol. One was treated for DCS elsewhere. The protocol included: pretreatment evaluation; chest radiograph; spirometry; blood gases; a single, 2-atmospheres absolute (atm abs), 120-minute HBO2 treatment; and a gradual acceleration over one week to a twice-daily schedule contingent on clinical and laboratory findings. Bleomycin indications were: head-and-neck squamous cell carcinomas (11), Hodgkin's lymphoma (2), other carcinomas (2). HBO2 indications were: osteoradionecrosis (10), soft-tissue radionecrosis (3), DCS (1) and a provocative oxygen toxicity test for a military aviator (1). Total bleomycin doses ranged from 40 to 225u/m2 (mean +/- SD, 105 +/- 57) given in conjunction with other chemotherapies and/or radiation. Radiation was 63.3 +/- 31.72 Gy (mean +/- SD), none to the chest with the exception of one patient treated for DCS elsewhere. Other chemotherapies included: vinblastine (11), methotrexate (11), CCNU (6) cisplatinum (7), dacarbazin (2), Adriamycin (1), and vincristine (1). Median age at time of HBO2 was 52 years (range 22-77). Median bleomycin-to-HBO2 latency was 34 months (range 1-279). Three patients received HBO2 within six months, and seven patients received HBO2 within two years of their last bleomycin exposure. There were no adverse pre-to-post HBO2 changes in: arterial blood gases, spirometry, chest radiograph findings or clinical reports. There were no persistent post-HBO2 pulmonary complications on follow-up. Post-HBO2 data were available for 40%, 53%, 87% and 100% of these parameters respectively. DISCUSSION: Bleomycin and oxygen can individually cause acute pulmonary toxicity. However, evidence for increased long-term susceptibility based on their synergy may be overstated.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Bleomicina/administração & dosagem , Doença da Descompressão/terapia , Oxigenoterapia Hiperbárica/métodos , Lesões por Radiação/terapia , Adulto , Idoso , Antibióticos Antineoplásicos/efeitos adversos , Antineoplásicos/administração & dosagem , Bleomicina/efeitos adversos , Contraindicações , Feminino , Humanos , Pulmão/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Neoplasias/tratamento farmacológico , Osteorradionecrose/terapia , Fatores de Tempo , Adulto Jovem
14.
Free Radic Biol Med ; 53(8): 1584-94, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22940620

RESUMO

Acute lung injury (ALI) initiates protective responses involving genes downstream of the Nrf2 (Nfe2l2) transcription factor, including heme oxygenase-1 (HO-1), which stimulates mitochondrial biogenesis and related anti-inflammatory processes. We examined mitochondrial biogenesis during Staphylococcus aureus pneumonia in mice and the effect of Nrf2 deficiency on lung mitochondrial biogenesis and resolution of lung inflammation. S. aureus pneumonia established by nasal insufflation of live bacteria was studied in mitochondrial reporter (mt-COX8-GFP) mice, wild-type (WT) mice, and Nrf2⁻/⁻ mice. Bronchoalveolar lavage, wet/dry ratios, real-time RT-PCR and Western analysis, immunohistochemistry, and fluorescence microscopy were performed on the lung at 0, 6, 24, and 48 h. The mice survived S. aureus inoculations at 5×108 CFU despite diffuse lung inflammation and edema, but the Nrf2⁻/⁻ lung showed increased ALI. In mt-COX8-GFP mice, mitochondrial fluorescence was enhanced in bronchial and alveolar type II (AT2) epithelial cells. WT mice displayed rapid HO-1 upregulation and lower proinflammatory TNF-α, IL-1ß, and CCL2 and, especially in AT2 cells, higher anti-inflammatory IL-10 and suppressor of cytokine signaling-3 than Nrf2⁻/⁻ mice. In the alveolar region, WT but not Nrf2⁻/⁻ mice showed strongly induced nuclear respiratory factor-1, PGC-1α, mitochondrial transcription factor-A, SOD2, Bnip3, mtDNA copy number, and citrate synthase. These findings indicate that S. aureus pneumonia induces Nrf2-dependent mitochondrial biogenesis in the alveolar region, mainly in AT2 cells. Absence of Nrf2 suppresses the alveolar transcriptional network for mitochondrial biogenesis and anti-inflammation, which worsens ALI. The findings link redox activation of mitochondrial biogenesis to ALI resolution.


Assuntos
Lesão Pulmonar Aguda/etiologia , Renovação Mitocondrial , Fator 2 Relacionado a NF-E2/fisiologia , Pneumonia Estafilocócica/complicações , Pneumonia/etiologia , Alvéolos Pulmonares/patologia , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Western Blotting , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Feminino , Humanos , Técnicas Imunoenzimáticas , Mediadores da Inflamação/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxirredução , Pneumonia/patologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/patologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia , Edema Pulmonar/metabolismo , Edema Pulmonar/microbiologia , Edema Pulmonar/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Staphylococcus aureus/patogenicidade , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
15.
Gastroenterology ; 143(5): 1319-1329.e11, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22885334

RESUMO

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.


Assuntos
Transdiferenciação Celular/genética , Regulação da Expressão Gênica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Células Estreladas do Fígado/metabolismo , Miofibroblastos/metabolismo , Transdução de Sinais/genética , Actinas/genética , Actinas/metabolismo , Animais , Ductos Biliares , Tetracloreto de Carbono , Células Cultivadas , Perfilação da Expressão Gênica , Glicólise/genética , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/enzimologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Ácido Láctico/metabolismo , Ligadura , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias , Miofibroblastos/enzimologia , Piruvato Quinase/genética , Piruvato Quinase/metabolismo , RNA Mensageiro/metabolismo , Ratos , Fatores de Tempo
16.
J Appl Physiol (1985) ; 113(8): 1243-52, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22898548

RESUMO

Decreased physical performance is a well-known consequence of rapid ascent to high altitude. Hypoxic pulmonary vasoconstriction (HPV) potentially limits cardiac output and systemic blood flow, thus preventing successful adaptation to rapid ascent. We hypothesized that pharmacological enhancement of the heart rate with theophylline, combined with reversal of HPV via endothelin blockade, could increase exercise performance at high altitude. Female Sprague-Dawley rats were treated with combinations of 1) theophylline, 2) the endothelin receptor antagonists sitaxsentan/ambrisentan, and/or 3) phosphodiesterase-5 inhibitor sildenafil and exposed to either a simulated high altitude (4,267 m) or 12% oxygen. Exercise capacity, peripheral blood flow, hemodynamics, and pulmonary leak were examined. Combination treatment with theophylline and endothelin blockade, but not with the respective single compounds, significantly prolonged run-to-fatigue time under simulated high altitude. No such efficacy was found when theophylline was combined with sildenafil. Neither theophylline nor sitaxsentan or their combination influenced breathing rates and hemoglobin oxygen saturation. Whereas under hypoxia, theophylline significantly increased muscular blood flow, and sitaxsentan increased tissue oxygenation, the combination improved both parameters but in a reduced manner. Under hypoxia, the combination treatment but not the single compounds significantly enhanced pulmonary arterial pressure compared with controls (13.1 ± 6.3 vs. 11.9 ± 5.2 mmHg), whereas mean arterial pressure remained unaffected. Pulmonary wet-to-dry weight ratios were unaffected by combination treatment. We conclude that concomitant dosing with a cardiac stimulant and endothelin antagonist can partially reverse loss of physical performance capacity under hypobaric hypoxia, independent from improving blood oxygen saturation.


Assuntos
Antagonistas dos Receptores de Endotelina , Hipóxia/tratamento farmacológico , Hipóxia/fisiopatologia , Condicionamento Físico Animal/fisiologia , Teofilina/farmacologia , Altitude , Animais , Pressão Arterial/efeitos dos fármacos , Pressão Arterial/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Sinergismo Farmacológico , Feminino , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/fisiologia , Hemoglobinas/metabolismo , Hipóxia/metabolismo , Isoxazóis/farmacologia , Oxigênio/metabolismo , Fenilpropionatos/farmacologia , Piperazinas/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiologia , Purinas/farmacologia , Piridazinas/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores de Endotelina/metabolismo , Circulação Renal/efeitos dos fármacos , Circulação Renal/fisiologia , Respiração/efeitos dos fármacos , Citrato de Sildenafila , Sulfonas/farmacologia , Análise e Desempenho de Tarefas , Tiofenos/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
17.
J Oral Maxillofac Surg ; 70(7): 1573-83, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22698292

RESUMO

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.


Assuntos
Antibacterianos/uso terapêutico , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/terapia , Desbridamento/métodos , Oxigenoterapia Hiperbárica , Idoso , Alendronato/efeitos adversos , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/tratamento farmacológico , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/cirurgia , Conservadores da Densidade Óssea/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Terapia Combinada , Difosfonatos/efeitos adversos , Feminino , Seguimentos , Gengiva/patologia , Humanos , Imidazóis/efeitos adversos , Masculino , Mieloma Múltiplo/tratamento farmacológico , Osteoporose/tratamento farmacológico , Manejo da Dor , Pamidronato , Estudos Prospectivos , Qualidade de Vida , Resultado do Tratamento , Cicatrização/fisiologia , Ácido Zoledrônico
19.
J Appl Physiol (1985) ; 112(11): 1814-23, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22442027

RESUMO

In hyperbaric oxygen (HBO(2)) at or above 3 atmospheres absolute (ATA), autonomic pathways link central nervous system (CNS) oxygen toxicity to pulmonary damage, possibly through a paradoxical and poorly characterized relationship between central nitric oxide production and sympathetic outflow. To investigate this possibility, we assessed sympathetic discharges, catecholamine release, cardiopulmonary hemodynamics, and lung damage in rats exposed to oxygen at 5 or 6 ATA. Before HBO(2) exposure, either a selective inhibitor of neuronal nitric oxide synthase (NOS) or a nonselective NOS inhibitor was injected directly into the cerebral ventricles to minimize effects on the lung, heart, and peripheral circulation. Experiments were performed on both anesthetized and conscious rats to differentiate responses to HBO(2) from the effects of anesthesia. EEG spikes, markers of CNS toxicity in anesthetized animals, were approximately four times as likely to develop in control rats than in animals with central NOS inhibition. In inhibitor-treated animals, autonomic discharges, cardiovascular pressures, catecholamine release, and cerebral blood flow all remained below baseline throughout exposure to HBO(2). In control animals, however, initial declines in these parameters were followed by significant increases above their baselines. In awake animals, central NOS inhibition significantly decreased the incidence of clonic-tonic convulsions or delayed their onset, compared with controls. The novel findings of this study are that NO produced by nNOS in the periventricular regions of the brain plays a critical role in the events leading to both CNS toxicity in HBO(2) and to the associated sympathetic hyperactivation involved in pulmonary injury.


Assuntos
Fibras Adrenérgicas/fisiologia , Sistema Nervoso Central/fisiopatologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/fisiopatologia , Óxido Nítrico/fisiologia , Oxigênio/toxicidade , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Fibras Adrenérgicas/efeitos dos fármacos , Animais , Sistema Nervoso Central/efeitos dos fármacos , Oxigenoterapia Hiperbárica/efeitos adversos , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/metabolismo , Oxigênio/administração & dosagem , Ratos , Ratos Sprague-Dawley
20.
Am J Respir Crit Care Med ; 185(8): 851-61, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22312014

RESUMO

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.


Assuntos
Monóxido de Carbono/farmacologia , Heme Oxigenase-1/metabolismo , Mitocôndrias/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Sepse/enzimologia , Infecções Estafilocócicas/terapia , Administração por Inalação , Animais , Western Blotting , Monóxido de Carbono/metabolismo , Modelos Animais de Doenças , Feminino , Heme Oxigenase-1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Fator 2 Relacionado a NF-E2/genética , Biogênese de Organelas , Peritonite/tratamento farmacológico , Peritonite/microbiologia , Peritonite/mortalidade , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Medição de Risco , Sepse/genética , Sepse/mortalidade , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Taxa de Sobrevida , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA