Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Article in English | MEDLINE | ID: mdl-27462149

ABSTRACT

INTRODUCTION: Severe chronic obstructive pulmonary disease (COPD) is often associated with secondary pulmonary hypertension (PH), which worsens prognosis. PH can be lowered by oxygen, but also by inhaled nitric oxide (NO), which has the potential to improve the health status of these patients. NO is an important mediator in vascular reactions in the pulmonary circulation. Oral compounds can act through NO-mediated pathways, but delivering pulsed inhaled NO (iNO) directly to the airways and pulmonary vasculature could equally benefit patients. Therefore, a proof-of-concept study was performed to quantify pulmonary blood vessel caliber changes after iNO administration using computed tomography (CT)-based functional respiratory imaging (FRI). METHODS: Six patients with secondary PH due to COPD received "pulsed" iNO in combination with oxygen for 20 minutes via a nasal cannula. Patients underwent a high-resolution CT scan with contrast before and after iNO. Using FRI, changes in volumes of blood vessels and associated lobes were quantified. Oxygen saturation and blood pressure were monitored and patients were asked about their subjective feelings. RESULTS: Pulmonary blood vessel volume increased by 7.06%±5.37% after iNO. A strong correlation (Ω(2) 0=0.32, P=0.002) was obtained between ventilation and observed vasodilation, suggesting that using the pulsed system, iNO is directed toward the ventilated zones, which consequently experience more vasodilation. Patients did not develop oxygen desaturation, remained normotensive, and perceived an improvement in their dyspnea sensation. CONCLUSION: Inhalation of pulsed NO with oxygen causes vasodilation in the pulmonary circulation of COPD patients, mainly in the well-ventilated areas. A high degree of heterogeneity was found in the level of vasodilation. Patients tend to feel better after the treatment. Chronic use trials are warranted.


Subject(s)
Antihypertensive Agents/administration & dosage , Hypertension, Pulmonary/drug therapy , Lung/blood supply , Nitric Oxide/administration & dosage , Oxygen Inhalation Therapy , Pulmonary Artery/drug effects , Pulmonary Disease, Chronic Obstructive/therapy , Vasodilation/drug effects , Vasodilator Agents/administration & dosage , Administration, Inhalation , Aged , Antihypertensive Agents/adverse effects , Arterial Pressure/drug effects , Computed Tomography Angiography , Female , Forced Expiratory Volume , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Lung/physiopathology , Male , Nitric Oxide/adverse effects , Oxygen Inhalation Therapy/adverse effects , Patient Satisfaction , Perfusion Imaging , Pulmonary Artery/diagnostic imaging , Pulmonary Artery/physiopathology , Pulmonary Circulation/drug effects , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulse Therapy, Drug , Time Factors , Treatment Outcome , Vasodilator Agents/adverse effects , Vital Capacity
2.
J Heart Lung Transplant ; 34(11): 1366-75, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26210752

ABSTRACT

BACKGROUND: Imatinib is an oral inhibitor of several protein kinases implicated in the pathophysiology of pulmonary hypertension. Treatment with imatinib resulted in improved hemodynamics and exercise capacity in a controlled trial (Imatinib [QTI571] in Pulmonary Arterial Hypertension, a Randomized Efficacy Study [IMPRES]), among pulmonary arterial hypertension (PAH) patients inadequately responsive to 2 to 3 PAH-specific therapies. METHODS: The long-term (up to 204 weeks) safety and efficacy of imatinib in this open-label extension study were reviewed until early study termination on April 16, 2014. Of 202 IMPRES-enrolled patients, 66 imatinib and 78 placebo recipients entered the extension. RESULTS: Overall, 93.8% (135 of 144) of patients discontinued the extension study; administrative issues (i.e., sponsor termination; 32.6%) and adverse events (31.3%) were the primary reasons for discontinuation. Nine patients completed the extension study before it was terminated. Serious and unexpected adverse events were frequent. These included 6 subdural hematomas in the extension study and 17 deaths during or within 30 days of study end. Although the patients who tolerated imatinib and remained in the extension for a longer duration did experience an improvement in functional class and walk distance, most discontinued the drug and the study. CONCLUSIONS: Severe adverse events, significant side effects, and a high discontinuation rate limit the utility of imatinib in the treatment of PAH. These risks outweigh any possible improvements in hemodynamics and walk distance seen in those patients able to remain on drug. The off-label use of this compound in PAH is discouraged.


Subject(s)
Hemodynamics/drug effects , Hypertension, Pulmonary/drug therapy , Imatinib Mesylate/administration & dosage , Adolescent , Adult , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Exercise Tolerance/physiology , Female , Follow-Up Studies , Humans , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Retrospective Studies , Time Factors , Treatment Outcome , Young Adult
3.
Tohoku J Exp Med ; 236(4): 255-61, 2015 08.
Article in English | MEDLINE | ID: mdl-26156407

ABSTRACT

Mechanical ventilation is the most important life supportive therapy for patients with acute respiratory distress syndrome (ARDS). However, increasing evidence from clinical studies suggests that mechanical ventilation can cause lung fibrosis, which may significantly contribute to morbidity and mortality. Recent studies also found fibroproliferation occurred in early stage of ARDS with poor outcome. We have hypothesized that mechanical ventilation-induced lung injury may be a major contributor to lung fibrosis, and antioxidant could be a potential therapeutic agent for the treatment to mechanic ventilation induced fibroproliferation. We therefore used Sprague-Dawley rats that were ventilated with large tidal volume (20 ml/kg) or low tidal volume (7 ml/kg). We analyzed the time course of collagen level in the lung and the effect of N-acetylcysteine (NAC), a thiol antioxidant, on mechanical ventilation-induced collagen accumulation. In addition, normal human lung fibroblasts (NHLF) were exposed to mechanical stretch, which mimics ventilator-induced lung inflation, to evaluate the collagen secretion in culture medium. We found that ventilation-induced collagen accumulation occurred even after 2-hour ventilation. Pretreatment with NAC (140 mg/kg) inhibited collagen accumulation in lungs of rats ventilated with large tidal volume. Moreover, mechanical stretch caused the accumulation of collagen in the culture medium of NHLF, the magnitude of which was decreased with the pretreatment with NAC (1 mM). These results indicate that mechanical ventilation can induce collagen accumulation within 2 hours. NAC alleviated the collagen accumulation induced by mechanical ventilation with high tidal volume. Therefore, NAC can be considered as a good candidate in preventing ventilation-induced lung fibrosis.


Subject(s)
Acetylcysteine/pharmacology , Collagen/metabolism , Lung/pathology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , Respiration, Artificial/adverse effects , Acetylcysteine/administration & dosage , Analysis of Variance , Animals , Collagen/drug effects , Injections, Intravenous , Lung/metabolism , Pulmonary Fibrosis/etiology , Rats , Rats, Sprague-Dawley , Tidal Volume , Time Factors
4.
Pulm Circ ; 4(3): 452-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25621158

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by severe remodeling of the pulmonary artery resulting in increased pulmonary artery pressure and right ventricular hypertrophy and, ultimately, failure. Bone marrow-derived progenitor cells play a critical role in vascular homeostasis and have been shown to be involved in the pathogenesis of PAH. A proliferation of c-Kit(+) hematopoietic progenitors and mast cells has been noted in the remodeled vessels in PAH. Imatinib, a tyrosine kinase inhibitor that targets c-Kit, has been shown to be beneficial for patients with PAH. Here we hypothesize that the clinical benefit of imatinib in PAH could be related to c-Kit inhibition of progenitor cell mobilization and maturation into mast cells. As a corollary to the phase 3 study using imatinib in PAH, blood samples were collected from 12 patients prior to starting study drug (baseline) and while on treatment at weeks 4 and 24. Eight were randomized to imatinib and 4 to placebo. Circulating c-Kit(+) and CD34(+)CD133(+) hematopoietic progenitors as well as biomarkers of mast cell numbers and activation were measured. Circulating CD34(+)CD133(+) and c-Kit(+) progenitor cells as well as c-Kit(+)/CD34(+)CD133(+) decreased with imatinib therapy (all P < 0.05). In addition, total tryptase, a marker of mast cell load, dropped with imatinib therapy (P = 0.02) and was related to pulmonary vascular resistance (R = 0.7, P = 0.02). The findings support c-Kit inhibition as a potential mechanism of action of imatinib in PAH and suggest that tryptase is a potential biomarker of response to therapy.

5.
Circulation ; 127(10): 1128-38, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23403476

ABSTRACT

BACKGROUND: By its inhibitory effect on platelet-derived growth factor signaling, imatinib could be efficacious in treating patients with pulmonary arterial hypertension (PAH). METHODS AND RESULTS: Imatinib in Pulmonary Arterial Hypertension, a Randomized, Efficacy Study (IMPRES), a randomized, double-blind, placebo-controlled 24-week trial, evaluated imatinib in patients with pulmonary vascular resistance ≥ 800 dyne·s·cm(-5) symptomatic on ≥ 2 PAH therapies. The primary outcome was change in 6-minute walk distance. Secondary outcomes included changes in hemodynamics, functional class, serum levels of N-terminal brain natriuretic peptide, and time to clinical worsening. After completion of the core study, patients could enter an open-label long-term extension study. Of 202 patients enrolled, 41% patients received 3 PAH therapies, with the remainder on 2 therapies. After 24 weeks, the mean placebo-corrected treatment effect on 6-minute walk distance was 32 m (95% confidence interval, 12-52; P=0.002), an effect maintained in the extension study in patients remaining on imatinib. Pulmonary vascular resistance decreased by 379 dyne·s·cm(-5) (95% confidence interval, -502 to - 255; P<0.001, between-group difference). Functional class, time to clinical worsening, and mortality did not differ between treatments. Serious adverse events and discontinuations were more frequent with imatinib than placebo (44% versus 30% and 33% versus 18%, respectively). Subdural hematoma occurred in 8 patients (2 in the core study, 6 in the extension) receiving imatinib and anticoagulation. CONCLUSIONS: Imatinib improved exercise capacity and hemodynamics in patients with advanced PAH, but serious adverse events and study drug discontinuations were common. Further studies are needed to investigate the long-term safety and efficacy of imatinib in patients with PAH. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00902174 (core study); NCT01392495 (extension).


Subject(s)
Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Piperazines/administration & dosage , Piperazines/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Adolescent , Adult , Aged , Benzamides , Double-Blind Method , Exercise Tolerance/drug effects , Exercise Tolerance/physiology , Familial Primary Pulmonary Hypertension , Female , Hematoma, Subdural/chemically induced , Hematoma, Subdural/enzymology , Hematoma, Subdural/physiopathology , Humans , Hypertension, Pulmonary/enzymology , Imatinib Mesylate , Male , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Young Adult
6.
Swiss Med Wkly ; 141: w13255, 2011.
Article in English | MEDLINE | ID: mdl-21989944

ABSTRACT

QUESTIONS UNDER STUDY: New evidence demonstrated that high tidal volume mechanical ventilation results in substantial bronchial airway mechanical strain. In addition, high tidal volume mechanical ventilation has been shown to increase IL-8 production in a mechanism mediated, at least in part, by low molecular weight hyaluronan (LWM-HA). In the present study, it was investigated whether LMW-HA synthesised in the lung, in response to cyclic stretch, increased IL-8 production in the bronchial epithelium. METHODS: This question was approached by stimulating a transformed human bronchial epithelial cell line with LMW-HA isolated from stretched human lung fibroblasts and probed for the activation of extracellular signal-regulated kinase pathways. RESULTS: LMW-HA increased IL-8 secretion in transformed bronchial epithelial cells. Additionally, LMW-HA augmented the levels of phospho c-Jun NH2-terminal kinase (JNK) and phospho extracellular signal-regulated kinase 1/2 (ERK1/2), and also mobilised nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) from the cytoplasm to the nucleus. The inhibition of JNK, ERK1/2 and NF-κB blocked IL-8 secretion in response to LMW-HA. CONCLUSION: The data suggest that LMW-HA produced by lung fibroblasts in response to cyclic stretch increases the secretion of IL-8 in transformed bronchial epithelial cells via AP-1 and NF-κB signalling pathways. These findings support the hypothesis that LMW-HA plays an active role in acute lung inflammation triggered by mechanical strain.


Subject(s)
Bronchi/injuries , Epithelial Cells/drug effects , Hyaluronic Acid/physiology , Interleukin-8/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Epithelial Cells/metabolism , Humans , Respiration, Artificial/adverse effects , Tidal Volume/physiology
7.
J Immunol ; 186(1): 556-62, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098223

ABSTRACT

Intratracheal administration of low molecular mass (LMM) hyaluronan (200 kDa) results in greater neutrophil infiltration in the lungs of TLR4(-/-) mice compared with that in wild-type mice. In general, enhanced neutrophil infiltration in tissue is due to cell influx; however, neutrophil apoptosis also plays an important role. We have assessed the effects of TLR4 in the regulation of neutrophil apoptosis in response to administration of LMM hyaluronan. We found that apoptosis of inflammatory neutrophils is impaired in TLR4(-/-) mice, an effect that depends upon the IFN-ß-mediated TRAIL/TRAILR system. IFN-ß levels were decreased in LMM hyaluronan-treated TLR4-deficient neutrophils. The treatment of inflammatory neutrophils with IFN-ß enhanced the levels of TRAIL and TRAILR 2. LMM hyaluronan-induced inflammatory neutrophil apoptosis was substantially prevented by anti-TRAIL neutralizing mAb. We conclude that decreased IFN-ß levels decrease the activity of the TRAIL/TRAILR system in TLR4-deficient neutrophils, leading to impaired apoptosis of neutrophils and resulting in abnormal accumulation of neutrophils in the lungs of LMM hyaluronan-treated mice. Thus, TLR4 plays a novel homeostatic role in noninfectious lung inflammation by accelerating the elimination of inflammatory neutrophils.


Subject(s)
Apoptosis/immunology , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/physiology , Interferon-beta/physiology , Neutrophils/immunology , Toll-Like Receptor 4/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Homeostasis/immunology , Hyaluronic Acid/chemistry , Inflammation Mediators/administration & dosage , Inflammation Mediators/chemistry , Inflammation Mediators/physiology , Interferon-beta/deficiency , Interferon-beta/genetics , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Weight , Neutrophil Activation/drug effects , Neutrophil Activation/immunology , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Neutrophils/drug effects , Neutrophils/pathology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
8.
Am J Respir Cell Mol Biol ; 44(4): 524-30, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20558775

ABSTRACT

Ras homolog gene family member A (RhoA) through Rho kinase kinase (ROCK), one of its downstream effectors, regulates a wide range of cell physiological functions, including vascular smooth muscle cell (SMC) proliferation, by degrading cyclin-dependent kinase inhibitor, p27. Our previous studies found that heparin inhibition of pulmonary artery SMC (PASMC) proliferation and pulmonary hypertension was dependent on p27 up-regulation. To investigate whether ROCK, a regulator of p27, is involved in regulation of heparin inhibition of PASMC proliferation, we analyzed ROCK expression in the lungs from mice and from human PASMCs exposed to hypoxia, and investigated the effect of ROCK expression in vitro by RhoA cDNA transfection. We also investigated the effect of guanine nucleotide exchange factor (GEF)-H1, an upstream regulator of RhoA, on heparin inhibition of PASMC proliferation by GEF-H1 cDNA transfection. We found that: (1) hypoxia increased ROCK expression in mice and PASMCs; (2) overexpression of RhoA diminished the inhibitory effect of heparin on PASMC proliferation and down-regulated p27 expression; and (3) overexpression of GEF-H1 negated heparin inhibition of PASMC proliferation, which was accompanied by increased GTP-RhoA and decreased p27. This study demonstrates that the RhoA/ROCK pathway plays an important role in heparin inhibition on PASMC proliferation, and reveals that heparin inhibits PASMC proliferation through GEF-H1/RhoA/ROCK/p27 signaling pathway, by down-regulating GEF-H1, RhoA, and ROCK, and then up-regulating p27.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Heparin/pharmacology , Myocytes, Smooth Muscle/cytology , Pulmonary Artery/cytology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cattle , Cell Cycle/drug effects , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Guanine Nucleotide Exchange Factors/genetics , Guanosine Triphosphate/metabolism , Humans , Mice , Models, Biological , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , RNA, Small Interfering/metabolism , Rho Guanine Nucleotide Exchange Factors
9.
Respir Res ; 11: 172, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21134294

ABSTRACT

BACKGROUND: Increased mucus secretion is one of the important characteristics of the response to smoke inhalation injuries. We hypothesized that gel-forming mucins may contribute to the increased mucus production in a smoke inhalation injury. We investigated the role of c-Jun N-terminal kinase (JNK) in modulating smoke-induced mucus secretion. METHODS: We intubated mice and exposed them to smoke from burning cotton for 15 min. Their lungs were then isolated 4 and 24 h after inhalation injury. Three groups of mice were subjected to the smoke inhalation injury: (1) wild-type (WT) mice, (2) mice lacking JNK1 (JNK1-/- mice), and (3) WT mice administered a JNK inhibitor. The JNK inhibitor (SP-600125) was injected into the mice 1 h after injury. RESULTS: Smoke exposure caused an increase in the production of mucus in the airway epithelium of the mice along with an increase in MUC5AC gene and protein expression, while the expression of MUC5B was not increased compared with control. We found increased MUC5AC protein expression in the airway epithelium of the WT mice groups both 4 and 24 h after smoke inhalation injury. However, overproduction of mucus and increased MUC5AC protein expression induced by smoke inhalation was suppressed in the JNK inhibitor-treated mice and the JNK1 knockout mice. Smoke exposure did not alter the expression of MUC1 and MUC4 proteins in all 3 groups compared with control. CONCLUSION: An increase in epithelial MUC5AC protein expression is associated with the overproduction of mucus in smoke inhalation injury, and that its expression is related on JNK1 signaling.


Subject(s)
Lung/metabolism , MAP Kinase Kinase 4/metabolism , Mucus/metabolism , Smoke Inhalation Injury/metabolism , Animals , Enzyme Activation , Mice , Mice, Inbred C57BL
10.
Respirology ; 15(7): 1131-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20874749

ABSTRACT

BACKGROUND AND OBJECTIVE: High MW hyaluronan (HMW HA) as opposed to low MW hyaluronan (LMW HA) has been shown to have anti-inflammatory and anti-apoptotic effects. We hypothesized that treatment with HMW HA would block smoke inhalation lung injury by inhibiting smoke-induced lung inflammation and airway epithelial cell apoptosis. METHODS: Anesthetized, intubated male rats were randomly allocated to either control or smoke inhalation injury groups. Rats were treated with 3-mL subcutaneous normal saline solution (sham) or LMW HA (35 kDa) or HMW HA (1600 kDa) 18 h before exposure to 15 min of cotton smoke (n = 5 each). Rats were also treated post smoke inhalation with 1600 kDa HA by intra-peritoneal injection (3 mL) or intra-tracheal nebulization (200 µL). Lung neutrophil infiltration, airway apoptosis, airway mucous plugging and lung injury were assessed 4 h after smoke inhalation injury. RESULTS: Rats pretreated with 1600 kDa HA had significantly less smoke-induced neutrophil infiltration, lung oedema, airway apoptosis and mucous plugging. Pretreatment with 35 kDa HA, in contrast, increased smoke-induced neutrophil infiltration and lung injury score. Intra-tracheal administration of a single dose 1600 kDa HA, but not intra-peritoneal injection, significantly improved survival post smoke inhalation. CONCLUSIONS: High MW hyaluronan (1600 kDa) may prove to be a beneficial therapy for smoke inhalation through inhibition of smoke-induced inflammation, lung oedema, airway epithelial cell apoptosis and airway mucous plugging.


Subject(s)
Hyaluronic Acid/therapeutic use , Smoke Inhalation Injury/drug therapy , Animals , Apoptosis/drug effects , Lung/drug effects , Lung/immunology , Lung Injury/drug therapy , Male , Mucus/drug effects , Mucus/immunology , Neutrophil Infiltration/drug effects , Pneumonia/drug therapy , Pulmonary Edema/drug therapy , Rats , Rats, Sprague-Dawley
11.
J Cardiothorac Surg ; 5: 32, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20441584

ABSTRACT

BACKGROUND AND OBJECTIVE: Chronic hypoxia induces pulmonary hypertension in mice. Smooth muscle cell hyperplasia and medial thickening characterize the vasculature of these animals. Thrombospondin-1 null (TSP-1(-/-)) mice spontaneously develop pulmonary smooth muscle cell hyperplasia and medial thickening. In addition, TSP-1 produced by the pulmonary endothelium inhibits pulmonary artery smooth muscle cell growth. Based on these observations we sought to describe the pulmonary vascular changes in TSP-1(-/-) mice exposed to chronic hypoxia. METHODS: We exposed TSP-1(-/-) and wild type (WT) mice to a fraction of inspired oxygen (FiO2) of 0.1 for up to six weeks. Pulmonary vascular remodeling was evaluated using tissue morphometrics. Additionally, right ventricle systolic pressures (RVSP) and right ventricular hypertrophy by right ventricle/left ventricle + septum ratios (RV/LV+S) were measured to evaluate pulmonary hypertensive changes. Finally, acute pulmonary vasoconstriction response in both TSP-1(-/-) and WT mice was evaluated by acute hypoxia and U-46619 (a prostaglandin F2 analog) response. RESULTS: In hypoxia, TSP-1(-/-) mice had significantly lower RVSP, RV/LV+S ratios and less pulmonary vascular remodeling when compared to WT mice. TSP-1(-/-) mice also had significantly lower RVSP in response to acute pulmonary vasoconstriction challenges than their WT counterparts. CONCLUSION: TSP-1(-/-) mice had diminished pulmonary vasoconstriction response and were less responsive to hypoxia-induced pulmonary hypertension than their wild type counterparts. This observation suggests that TSP-1 could play an active role in the pathogenesis of pulmonary hypertension associated with hypoxia.


Subject(s)
Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Thrombospondin 1/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/physiopathology , Thrombospondin 1/genetics , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Ventricular Function, Right
12.
J Immunol ; 184(9): 5308-14, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20357263

ABSTRACT

Low m.w. hyaluronan (LMW HA) has been shown to elicit the expression of proinflammatory cytokines and chemokines in various cells in vitro. However, the effects of this molecule in vivo are unknown. In this study, we report that intratracheal administration of LMW HA (200 kDa) causes inflammation in mouse lung. A lack of TLR4 is associated with even stronger inflammatory response in the lung as shown by increased neutrophil counts and elevated cytokine and chemokine concentrations. We also demonstrate that TLR4 anti-inflammatory signaling is dependent upon a MyD88-independent pathway. TLR4-mediated IL-1R antagonist production plays a negative regulatory role in LMW HA (200 kDa) induced lung inflammation. These data provide a molecular level explanation for the function of TLR4 in LMW HA (200 kDa)-induced lung inflammation, as inhibition of the beta form of pro-IL-1 promotes an anti-inflammatory response.


Subject(s)
Acute Lung Injury/immunology , Acute Lung Injury/pathology , Down-Regulation/immunology , Inflammation Mediators/physiology , Lung/immunology , Lung/pathology , Toll-Like Receptor 4/physiology , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Membrane Permeability/immunology , Cells, Cultured , Down-Regulation/genetics , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/chemistry , Inflammation Mediators/metabolism , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin 1 Receptor Antagonist Protein/physiology , Interleukin-1beta/antagonists & inhibitors , Lung/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Molecular Weight , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Signal Transduction/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
13.
Transl Res ; 154(5): 228-40, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19840764

ABSTRACT

High-tidal-volume mechanical ventilation and hyperoxia used in patients with acute lung injury (ALI) can induce alveolar coagulopathy and fibrin depositions within the airways. Hyperoxia has been shown to increase ventilator-induced lung injury (VILI), but the mechanisms that regulate interaction between high-tidal-volume mechanical ventilation and hyperoxia are unclear. We hypothesized that mechanical stretch with hyperoxia synergistically augmented neutrophil infiltration and production of plasminogen activator inhibitor-1 (PAI-1) via the nuclear factor-kappaB (NF-kappaB) pathway. C57BL/6 mice (n=5 per group) were exposed to high-tidal-volume (30 mL/kg) or low-tidal-volume (6 mL/kg) mechanical ventilation with room air or hyperoxia for 1 to 5h after 2-microg/g NF-kappaB inhibitor (SN-50) administration. Nonventilated mice with room air or hyperoxia served as control groups. Evans blue dye, myeloperoxidase, electrophoretic mobility shifting of nuclear protein, and inflammatory cytokine were measured. The expression of tumor necrosis factor-alpha (TNF-alpha) and PAI-1 were studied by immunohistochemistry. The addition of hyperoxia to high-tidal-volume ventilation-augmented lung injury, as demonstrated by increased microvascular leak, neutrophil migration into the lung, TNF-alpha and active PAI-1 production, DNA binding activity of NF-kappaB, and NF-kappaB activation. No statistically significant increase of neutrophil infiltration and inflammatory cytokine production was found in the mice ventilated at 6 mL/kg using hyperoxia. Hyperoxia-induced augmentation of VILI was attenuated in mice with pharmacologic inhibition of NF-kappaB activity by SN-50. We conclude that hyperoxia increased high-tidal-volume-induced cytokine production and neutrophil influx through activation of the NF-kappaB pathway.


Subject(s)
Acute Lung Injury/metabolism , Disseminated Intravascular Coagulation/etiology , Hyperoxia/complications , NF-kappa B/metabolism , Respiration, Artificial/adverse effects , Acute Lung Injury/pathology , Animals , DNA/metabolism , Disease Models, Animal , Disseminated Intravascular Coagulation/metabolism , Hyperoxia/metabolism , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Peptides/toxicity , Protein Binding , Respiration, Artificial/methods , Serpin E2 , Serpins/metabolism , Tidal Volume
14.
Crit Care ; 13(4): R108, 2009.
Article in English | MEDLINE | ID: mdl-19580651

ABSTRACT

INTRODUCTION: Dysregulation of coagulation and local fibrinolysis found in patients with acute lung injury often results in the need for the support of mechanical ventilation. High-tidal-volume mechanical ventilation can increase lung damage and suppression of fibrinolytic activity, but the mechanisms are unclear. We hypothesized that subcutaneous injections of unfractionated heparin and enoxaparin would decrease neutrophil infiltration, lung edema, and plasminogen-activator inhibitor-1 (PAI-1) production in mice exposed to high-tidal-volume ventilation. METHODS: Male C57BL/6 mice, weighing 20 to 25 g, were exposed to either high-tidal-volume (30 ml/kg) or low-tidal-volume (6 ml/kg) mechanical ventilation with room air for 1 to 5 hours after 200 IU/kg or 400 IU/kg unfractionated heparin and 4 mg/kg or 8 mg/kg enoxaparin administration. Nonventilated mice served as a control group. Evan blue dye, lung wet- to dry-weight ratio, histopathologic grading of epithelium, myeloperoxidase, and gene expression of PAI-1 were measured. The expression of PAI-1 was studied by immunohistochemistry. RESULTS: High-tidal-volume ventilation induced increased microvascular permeability, neutrophil influx, PAI-1 mRNA expression, production of PAI-1 protein, and positive staining of PAI-1 in epithelium in a dose-dependent manner. Lung injury induced by high-tidal-volume ventilation was attenuated with PAI-1-deficient mice and pharmacologic inhibition of PAI-1 activity by low-dose unfractionated heparin and enoxaparin. CONCLUSIONS: We conclude that high-tidal-volume mechanical ventilation increased microvascular permeability, neutrophil influx, lung PAI-1 mRNA expression, production of active PAI-1. The deleterious effects were attenuated by low-dose unfractionated heparin or enoxaparin treatment. Understanding the protective mechanism of unfractionated heparin and enoxaparin related to the reduction of PAI-1 may afford further knowledge of the effects of mechanical forces in the lung and development of possible therapeutic strategies involved in acute lung injury.


Subject(s)
Anticoagulants/therapeutic use , Enoxaparin/therapeutic use , Heparin/therapeutic use , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/prevention & control , Animals , Base Sequence , Body Water , DNA Primers , Enoxaparin/pharmacology , Heparin/pharmacology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Plasminogen Activator Inhibitor 1/genetics , Prospective Studies , RNA, Messenger/genetics , Respiration, Artificial/methods , Reverse Transcriptase Polymerase Chain Reaction , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology
15.
Lung ; 187(4): 233-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19572173

ABSTRACT

Low-molecular-weight hyaluronan produced by hyaluronan synthase 3 (HAS3) has been shown to play a role in acute lung injury secondary to high-tidal-volume ventilation. Phosphodiesterase 3 inhibitors have been shown to decrease HAS3 expression. We hypothesized that low-molecular-weight hyaluronan (LMW HA) produced by HAS3 mediates LPS-induced lung injury in the mechanically ventilated rat and that milrinone (MIL), by blocking HAS3 mRNA expression, would prevent the injury. Rats were randomized to four groups: controls with mechanical ventilation at 7 cc/kg MV, MV+LPS, MV+MIL, and MV+LPS+MIL. Rats were intubated and ventilated without PEEP for 4 h. Lipopolysaccharide (LPS) (1 mg/kg) was infused into the arterial line 1 h prior to MV. MIL 10 microg/kg/min (or an equivalent volume of saline) was infused through the venous line at the beginning of MV. Bronchoalveolar lavage fluid (BAL) was collected after 4 h of ventilation and lungs were saved for histopathology. LPS significantly increased neutrophil infiltration and protein concentration in the BAL and augmented lung injury score on histology. MIL significantly lowered alveolar protein and neutrophil infiltration as well as lung injury in response to LPS. Furthermore, MIL decreased the mRNA expression for HAS3 and MIP2 in lung tissue and decreased the protein content in BAL. MIL, a commonly used inotropic agent, inhibited LPS-induced lung inflammation and lung injury in mechanically ventilated rats. The anti-inflammatory properties of MIL may be mediated by inhibition of HAS3 and/or MIP2 and could be beneficial in the treatment of sepsis.


Subject(s)
Acute Lung Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Glucuronosyltransferase/metabolism , Lung/drug effects , Milrinone/pharmacology , Phosphodiesterase 3 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Respiration, Artificial/adverse effects , Sepsis/drug therapy , Acute Lung Injury/enzymology , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Acute Lung Injury/physiopathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Capillary Permeability/drug effects , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Enzymologic/drug effects , Glucuronosyltransferase/genetics , Hemodynamics/drug effects , Hyaluronan Synthases , Hyaluronic Acid/metabolism , Lipopolysaccharides , Lung/enzymology , Lung/pathology , Male , Neutrophil Infiltration/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sepsis/complications , Sepsis/enzymology , Sepsis/pathology , Sepsis/physiopathology
16.
BMC Infect Dis ; 9: 80, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19486524

ABSTRACT

BACKGROUND: Acute kidney injury frequently accompanies sepsis. Endotoxin is known to reduce tissue levels of cAMP and low levels of cAMP have been associated with renal injury. We, therefore, hypothesized that endotoxin induced renal injury by activating phosphodiesterase 3 (PDE3) which metabolizes cAMP and that amrinone an inhibitor of PDE3 would prevent the renal injury. METHODS: Animals were divided into three groups (n = 7/group): 1) Control (0.9% NaCl infusion without LPS); 2) LPS (0.9% NaCl infusion with LPS); 3) Amrinone+LPS (Amrinone infusion with LPS). Either lipopolysaccharide (LPS) or vehicle was injected via the jugular vein and the rats followed for 3 hours. We explored the expression of PDE3 isoenzymes and the concentrations of cAMP in the tissue. RESULTS: The PDE3B gene but not PDE3A was upregulated in the kidney of LPS group. Immunohistochemistry also showed that PDE3B was expressed in the distal tubule in the controls and LPS caused PDE3B expression in the proximal as well. However, PDE3A was not expressed in the kidney either in the control or LPS treated groups. Tissue level of cAMP was decreased after LPS and was associated with an increase in blood urea nitrogen, creatinine, ultrastructural proximal tubular changes, and expression of inducible nitric oxide synthase (iNOS) in the endotoxemic kidney. In septic animals the phosphodiesterase 3 inhibitor, amrinone, preserved the tissue cAMP level, renal structural changes, and attenuated the increased blood urea nitrogen, creatinine, and iNOS expression in the kidney. CONCLUSION: These findings suggest a significant role for PDE3B as an important mediator of LPS-induced acute kidney injury.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Kidney/enzymology , Kidney/pathology , Lipopolysaccharides/toxicity , Amrinone/pharmacology , Animals , Blood Urea Nitrogen , Creatinine/metabolism , Cyclic AMP/metabolism , Gene Expression Regulation, Enzymologic , Kidney/metabolism , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/ultrastructure , Male , Nitric Oxide Synthase Type II/genetics , Rats
17.
Crit Care ; 12(4): R102, 2008.
Article in English | MEDLINE | ID: mdl-18691420

ABSTRACT

INTRODUCTION: Mechanical ventilation with even moderate-sized tidal volumes synergistically increases lung injury in sepsis and has been associated with proinflammatory low-molecular-weight hyaluronan production. High-molecular-weight hyaluronan (HMW HA), in contrast, has been found to be anti-inflammatory. We hypothesized that HMW HA would inhibit lung injury associated with sepsis and mechanical ventilation. METHODS: Sprague-Dawley rats were randomly divided into four groups: nonventilated control rats; mechanical ventilation plus lipopolysaccharide (LPS) infusion as a model of sepsis; mechanical ventilation plus LPS with HMW HA (1,600 kDa) pretreatment; and mechanical ventilation plus LPS with low-molecular-weight hyaluronan (35 kDa) pretreatment. Rats were mechanically ventilated with low (7 ml/kg) tidal volumes. LPS (1 or 3 mg/kg) or normal saline was infused 1 hour prior to mechanical ventilation. Animals received HMW HA or low-molecular-weight hyaluronan via the intraperitoneal route 18 hours prior to the study or received HMW HA (0.025%, 0.05% or 0.1%) intravenously 1 hour after injection of LPS. After 4 hours of ventilation, animals were sacrificed and the lung neutrophil and monocyte infiltration, the cytokine production, and the lung pathology score were measured. RESULTS: LPS induced lung neutrophil infiltration, macrophage inflammatory protein-2 and TNFalpha mRNA and protein, which were decreased in the presence of both 1,600 kDa and 35 kDa hyaluronan pretreatment. Only 1,600 kDa hyaluronan completely blocked both monocyte and neutrophil infiltration and decreased the lung injury. When infused intravenously 1 hour after LPS, 1,600 kDa hyaluronan inhibited lung neutrophil infiltration, macrophage inflammatory protein-2 mRNA expression and lung injury in a dose-dependent manner. The beneficial effects of hyaluronan were partially dependent on the positive charge of the compound. CONCLUSIONS: HMW HA may prove to be an effective treatment strategy for sepsis-induced lung injury with mechanical ventilation.


Subject(s)
Hyaluronic Acid/therapeutic use , Lung Injury/drug therapy , Respiration, Artificial/adverse effects , Sepsis/drug therapy , Animals , Drug Evaluation, Preclinical/methods , Hyaluronic Acid/chemistry , Lung Injury/etiology , Lung Injury/pathology , Rats , Rats, Sprague-Dawley , Sepsis/complications , Sepsis/pathology
18.
Crit Care ; 12(4): R103, 2008.
Article in English | MEDLINE | ID: mdl-18691424

ABSTRACT

INTRODUCTION: Lung fibrosis, reduced lung compliance, and severe hypoxemia found in patients with acute lung injury often result in a need for the support of mechanical ventilation. High-tidal-volume mechanical ventilation can increase lung damage and fibrogeneic activity but the mechanisms regulating the interaction between high tidal volume and lung fibrosis are unclear. We hypothesized that high-tidal-volume ventilation increased pulmonary fibrosis in acute lung injury via the serine/threonine kinase-protein kinase B (Akt) and mitogen-activated protein kinase pathways. METHODS: After 5 days of bleomycin administration to simulate acute lung injury, male C57BL/6 mice, weighing 20 to 25 g, were exposed to either high-tidal-volume mechanical ventilation (30 ml/kg) or low-tidal-volume mechanical ventilation (6 ml/kg) with room air for 1 to 5 hours. RESULTS: High-tidal-volume ventilation induced type I and type III procollagen mRNA expression, microvascular permeability, hydroxyproline content, Masson's trichrome staining, S100A4/fibroblast specific protein-1 staining, activation of Akt and extracellular signal-regulated kinase (ERK) 1/2, and production of macrophage inflammatory protein-2 and 10 kDa IFNgamma-inducible protein in a dose-dependent manner. High-tidal-volume ventilation-induced lung fibrosis was attenuated in Akt-deficient mice and in mice with pharmacologic inhibition of ERK1/2 activity by PD98059. CONCLUSION: We conclude that high-tidal-volume ventilation-induced microvascular permeability, lung fibrosis, and chemokine production were dependent, in part, on activation of the Akt and ERK1/2 pathways.


Subject(s)
Acute Lung Injury/enzymology , Bleomycin/toxicity , Extracellular Signal-Regulated MAP Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Pulmonary Fibrosis/enzymology , Respiration, Artificial/adverse effects , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Male , Mice , Mice, Inbred C57BL , Prospective Studies , Protein Serine-Threonine Kinases/physiology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology
19.
J Korean Med Sci ; 23(3): 406-13, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18583875

ABSTRACT

Atelectasis can impair arterial oxygenation and decrease lung compliance. However, the effects of atelectasis on endotoxemic lungs during ventilation have not been well studied. We hypothesized that ventilation at low volumes below functional residual capacity (FRC) would accentuate lung injury in lipopolysaccharide (LPS)-pretreated rats. LPS-pretreated rats were ventilated with room air at 85 breaths/min for 2 hr at a tidal volume of 10 mL/kg with or without thoracotomy. Positive end-expiratory pressure (PEEP) was applied to restore FRC in the thoracotomy group. While LPS or thoracotomy alone did not cause significant injury, the combination of endotoxemia and thoracotomy caused significant hypoxemia and hypercapnia. The injury was observed along with a marked accumulation of inflammatory cells in the interstitium of the lungs, predominantly comprising neutrophils and mononuclear cells. Immunohistochemistry showed increased inducible nitric oxide synthase (iNOS) expression in mononuclear cells accumulated in the interstitium in the injury group. Pretreatment with PEEP or an iNOS inhibitor (1400 W) attenuated hypoxemia, hypercapnia, and the accumulation of inflammatory cells in the lung. In conclusion, the data suggest that atelectasis induced by thoracotomy causes lung injury during mechanical ventilation in endotoxemic rats through iNOS expression.


Subject(s)
Endotoxemia/complications , Positive-Pressure Respiration/adverse effects , Pulmonary Atelectasis/etiology , Pulmonary Atelectasis/therapy , Thoracotomy/adverse effects , Animals , Blood Pressure , Carbon Dioxide/blood , Cardiac Output , Combined Modality Therapy , Endotoxemia/immunology , Endotoxemia/pathology , Functional Residual Capacity , Immunohistochemistry , Leukocytes, Mononuclear/pathology , Lipopolysaccharides/pharmacology , Lung/enzymology , Lung/pathology , Lung/physiopathology , Lung Compliance , Lung Volume Measurements , Male , Neutrophils/pathology , Nitric Oxide Synthase Type II/metabolism , Oxygen/blood , Pulmonary Atelectasis/pathology , Rats , Rats, Sprague-Dawley
20.
Respirology ; 13(3): 333-40, 2008 May.
Article in English | MEDLINE | ID: mdl-18399853

ABSTRACT

BACKGROUND AND OBJECTIVE: Ventilator-induced lung injury (VILI) leads to airway epithelial cell apoptosis and lung inflammation. High tidal volume ventilation in vivo has been shown to induce MIP-2 production, lung neutrophil sequestration and apoptotic airway cell death. This study aimed to determine the effect of N-acetylcysteine (NAC), a scavenger of oxygen radicals, on lung inflammation and apoptosis in an in vivo model of VILI. METHODS: Sprague-Dawley rats (n = 5 per group) were ventilated at low tidal volume (V(T) 7 mL/kg) or high tidal volume (V(T) 20 mL/kg) with or without administration of 140 mg/kg of intravenous NAC. Animals were ventilated for 30 min, 1 or 2 h, then allowed to recover for 2 h, at which time neutrophil infiltration, MIP-2, TNF-alpha and IL-6 in BAL fluid, as well as the percentage of apoptotic airway epithelial cells, were measured. RESULTS: Ventilation at V(T) 20 mL/kg increased oxidant release, as measured by serum isoprostane, and decreased lung glutathione, the major antioxidant in the lung. NAC treatment during ventilation at V(T) 20 mL/kg prevented the decrease in lung glutathione and significantly lowered serum isoprostane levels, neutrophil infiltration, cytokines in the BAL and apoptosis in the airways as compared with animals ventilated at V(T) 20 mL/kg without NAC (P < 0.05). CONCLUSIONS: These data point to an early role of oxidant-induced inflammation and apoptosis in VILI.


Subject(s)
Apoptosis/physiology , Oxidative Stress/physiology , Pneumonia/physiopathology , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/physiopathology , Ventilators, Mechanical/adverse effects , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid , Chemokine CXCL2/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Free Radical Scavengers/pharmacology , Glutathione/metabolism , Interleukin-6/metabolism , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Pneumonia/metabolism , Pneumonia/pathology , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...