ABSTRACT
BACKGROUND: In spite of the latest therapeutic developments, no effective treatments for handling critical conditions such as acute lung injuries have yet been found. Such conditions, which may result from lung infections, sepsis, multiple trauma, or shock, represent a significant challenge in intensive care medicine. Seeking ways to better deal with this challenge, the scientific community has recently devoted much attention to small molecules derived from natural products with anti-inflammatory and immunomodulatory effects. AIMS: In this context, we investigated the anti-inflammatory effect of Rubiadin-1-methyl ether isolated from Pentas schimperi, using an in vitro model of RAW 264.7 macrophages induced by LPS and an in vivo model of acute lung injury (ALI) induced by LPS. METHODS: The macrophages were pretreated with the compound and induced by LPS (1 µg/mL). After 24 h, using the supernatant, we evaluated the cytotoxicity, NOx, and IL-6, IL-1ß, and TNF-α levels, as well as the effect of the compound on macrophage apoptosis. Next, the compound was administered in mice with acute lung injury (ALI) induced by LPS (5 mg/kg), and the pro- and anti-inflammatory parameters were analyzed after 12 h using the bronchoalveolar lavage fluid (BALF). RESULTS: Rubiadin-1-methyl ether was able to inhibit the pro-inflammatory parameters studied in the in vitro assays (NOx, IL-6, and IL-1ß) and, at the same time, increased the macrophage apoptosis rate. In the in vivo experiments, this compound was capable of decreasing leukocyte infiltration; fluid leakage; NOx; IL-6, IL-12p70, IFN-γ, TNF-α, and MCP-1 levels; and MPO activity. In addition, Rubiadin-1-methyl ether increased the IL-10 levels in the bronchoalveolar lavage fluid (BALF). CONCLUSIONS: These findings support the evidence that Rubiadin-1-methyl ether has important anti-inflammatory activity, with evidence of an immunomodulatory effect.
Subject(s)
Anthraquinones/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Immunologic Factors/therapeutic use , Inflammation/drug therapy , Acute Lung Injury/blood , Acute Lung Injury/drug therapy , Animals , Cell Survival/drug effects , Inflammation/metabolism , Interleukin-1beta/blood , Interleukin-6/blood , Male , Mice , Nitric Oxide/metabolism , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/bloodABSTRACT
Purpose: The research is intended for clarification of the efficacy as well as the underlying mechanism of GSK-3β inhibitors on the advancement of acute lung injuries in acute necrotizing pancreatitis (ANP) in rats. Methods: Seventy-two rats were randomly divided into 6 groups: (1)ANP-vehicle; (2)ANP-TDZD-8;(3)ANP-SB216763;(4)Sham-vehicle;(5)Sham-TDZD-8;(6)Sham-SB216763; Blood biochemical test, histopathological examination and immunohistochemical analysis of rats pancreas and lung tissues were performed. The protein expression of GSK-3β, phospho-GSK-3β (Ser9), iNOS, ICAM-1, TNF-α, and IL-10 were detected in lung tissues by Western-blot. Results: The outcomes revealed that the intervention of GSK-3β inhibitors alleviated the pathological damage of pancreas and lung (P<0.01), reduced serum amylase, lipase, hydrothorax and lung Wet-to-Dry Ratio, attenuated serum concentrations of IL-1β and IL-6 (P<0.01), inhibited the activation of NF-κB, and abated expression of iNOS, ICAM-1 and TNF-α protein, but up-regulated IL-10 expression in lung of ANP rats (P<0.01). The inflammatory response and various indicators in ANP-TDZD-8 groups were lower than those in ANP-SB216763 groups. Conclusions: Inhibition of GSK-3β weakens acute lung injury related to ANP via the inhibitory function of NF-κB signaling pathway. Different kinds of GSK-3β inhibitors have different effects to ANP acute lung injury.(AU)
Subject(s)
Animals , Male , Rats , Acute Lung Injury/blood , Acute Lung Injury/classification , Acute Lung Injury/prevention & control , Acute Lung Injury/physiopathology , Pancreatitis, Acute Necrotizing/blood , Pancreatitis, Acute Necrotizing/complications , Pancreatitis, Acute Necrotizing/physiopathology , Glycogen Synthase Kinase 3 beta/administration & dosage , Glycogen Synthase Kinase 3 beta/analysis , China , Rats, WistarABSTRACT
PURPOSE: To investigate the apoptotic mechanisms in rabbits with blast-induced acute lung injury (ALI). METHODS: A total of 40 rabbits were randomly divided into a blank control group (A, n=10) and an experimental group (EXP, n=30). Explosion-induced chest-ALI models were prepared and sampled at different time points (4, 12, and 24h after modeling, T1-T3) to test the lung dry weight/wet weight ratio (W/D) and arterial oxygen pressure (PaO2), apoptosis of lung tissue by the TUNEL assay, and Caspase-3, Bax, and Bcl-2 levels by immunohistochemical analysis. Furthermore, lung tissue was sampled to observe pathological morphology by microscopy. RESULTS: Under a light microscope, Group EXP exhibited obvious edema in the pulmonary interstitial substance and alveoli, a large number of red blood cells, inflammatory cells, and serous exudation in the alveolar cavity, as well as thickening of the pulmonary interstitial fluid. Compared to Group A, the W/D ratio was significantly increased in Group EXP (P<0.01), while PaO2 was significantly reduced (P<0.01). The apoptosis index was significantly increased (P<0.01), and caspase-3 and Bax/Bcl-2 levels were increased (P<0.01). CONCLUSION: Apoptosis plays an important role in the occurrence and development of acute lung injury in rabbits by participating in lung injury and promoting the progression of ALI.
Subject(s)
Acute Lung Injury/physiopathology , Apoptosis/physiology , Blast Injuries/physiopathology , Acute Lung Injury/blood , Acute Lung Injury/pathology , Animals , Blast Injuries/blood , Blast Injuries/pathology , Caspase 3/blood , Disease Models, Animal , Female , Male , Proto-Oncogene Proteins c-bcl-2/blood , Pulmonary Alveoli/pathology , Rabbits , Random Allocation , bcl-2-Associated X Protein/bloodABSTRACT
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) affect >200 000 individuals yearly with a 40% mortality rate. Although platelets are implicated in the progression of ALI/ARDS, their exact role remains undefined. Triggering receptor expressed in myeloid cells (TREM)-like transcript 1 (TLT-1) is found on platelets, binds fibrinogen, and mediates clot formation. We hypothesized that platelets use TLT-1 to manage the progression of ALI/ARDS. Here we retrospectively measure plasma levels of soluble TLT-1 (sTLT-1) from the ARDS Network clinical trial and show that patients whose sTLT-1 levels were >1200 pg/mL had nearly twice the mortality risk as those with <1200 pg/mL (P < .001). After correcting for confounding factors such as creatinine levels, Acute Physiology And Chronic Health Evaluation III scores, age, platelet counts, and ventilation volume, sTLT-1 remains significant, suggesting that sTLT-1 is an independent prognostic factor (P < .0001). These data point to a role for TLT-1 during the progression of ALI/ARDS. We use a murine lipopolysaccharide-induced ALI model and demonstrate increased alveolar bleeding, aberrant neutrophil transmigration and accumulation associated with decreased fibrinogen deposition, and increased pulmonary tissue damage in the absence of TLT-1. The loss of TLT-1 resulted in an increased proportion of platelet-neutrophil conjugates (43.73 ± 24.75% vs 8.92 ± 2.4% in wild-type mice), which correlated with increased neutrophil death. Infusion of sTLT-1 restores normal fibrinogen deposition and reduces pulmonary hemorrhage by 40% (P ≤ .001) and tissue damage by 25% (P ≤ .001) in vivo. Our findings suggest that TLT-1 uses fibrinogen to govern the transition between inflammation and hemostasis and facilitate controlled leukocyte transmigration during the progression of ARDS.
Subject(s)
Acute Lung Injury/blood , Blood Platelets/metabolism , Receptors, Immunologic/blood , Respiratory Distress Syndrome/blood , Acute Lung Injury/pathology , Animals , Blood Platelets/pathology , Disease Models, Animal , Humans , Mice , Mice, Knockout , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Predictive Value of Tests , Respiratory Distress Syndrome/pathology , Transendothelial and Transepithelial MigrationABSTRACT
Abstract Purpose: To investigate the apoptotic mechanisms in rabbits with blast-induced acute lung injury (ALI). Methods: A total of 40 rabbits were randomly divided into a blank control group (A, n=10) and an experimental group (EXP, n=30). Explosion-induced chest-ALI models were prepared and sampled at different time points (4, 12, and 24h after modeling, T1-T3) to test the lung dry weight/wet weight ratio (W/D) and arterial oxygen pressure (PaO2), apoptosis of lung tissue by the TUNEL assay, and Caspase-3, Bax, and Bcl-2 levels by immunohistochemical analysis. Furthermore, lung tissue was sampled to observe pathological morphology by microscopy. Results: Under a light microscope, Group EXP exhibited obvious edema in the pulmonary interstitial substance and alveoli, a large number of red blood cells, inflammatory cells, and serous exudation in the alveolar cavity, as well as thickening of the pulmonary interstitial fluid. Compared to Group A, the W/D ratio was significantly increased in Group EXP (P<0.01), while PaO2 was significantly reduced (P<0.01). The apoptosis index was significantly increased (P<0.01), and caspase-3 and Bax/Bcl-2 levels were increased (P<0.01). Conclusion: Apoptosis plays an important role in the occurrence and development of acute lung injury in rabbits by participating in lung injury and promoting the progression of ALI.
Subject(s)
Animals , Male , Female , Rabbits , Blast Injuries/physiopathology , Apoptosis/physiology , Acute Lung Injury/physiopathology , Pulmonary Alveoli/pathology , Blast Injuries/pathology , Blast Injuries/blood , Random Allocation , Proto-Oncogene Proteins c-bcl-2/blood , Disease Models, Animal , bcl-2-Associated X Protein/blood , Caspase 3/blood , Acute Lung Injury/pathology , Acute Lung Injury/bloodABSTRACT
Sakuranetin is the main isolate flavonoid from Baccharis retusa (Asteraceae) leaves and exhibits anti-inflammatory and antioxidative activities. Acute respiratory distress syndrome is an acute failure of the respiratory system for which effective treatment is urgently necessary. This study investigated the preventive and therapeutic effects of sakuranetin on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. Animals were treated with intranasal sakuranetin 30 min before or 6 h after instillation of LPS. Twenty-four hours after ALI was induced, lung function, inflammation, macrophages population markers, collagen fiber deposition, the extent of oxidative stress, and the expression of matrix metalloprotease-9 (MMP-9), tissue inhibitor of MMP-9 (TIMP-1) and NF-κB were evaluated. The animals began to show lung alterations 6 h after LPS instillation, and these changes persisted until 24 h after LPS administration. Preventive and therapeutic treatment with sakuranetin reduced the neutrophils in the peripheral blood and in the bronchial alveolar lavage. Sakuranetin treatment also reduced macrophage populations, particularly that of M1-like macrophages. In addition, sakurnaetin treatment reduced keratinocyte-derived chemokines (IL-8 homolog) and NF-κB levels, collagen fiber formation, MMM-9 and TIMP-1-positive cells, and oxidative stress in lung tissues compared with LPS animals treated with vehicle. Finally, sakuranetin treatment also reduced total protein, and the levels of TNF-α and IL-1ß in the lung. This study shows that sakuranetin prevented and reduced pulmonary inflammation induced by LPS. Because sakuranetin modulates oxidative stress, the NF-κB pathway, and lung function, it may constitute a novel therapeutic candidate to prevent and treat ALI.
Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/prevention & control , Flavonoids/therapeutic use , Acute Lung Injury/blood , Acute Lung Injury/complications , Animals , Biomarkers/metabolism , Cell Polarity/drug effects , Collagen/metabolism , Compliance/drug effects , Cytokines/metabolism , Flavonoids/chemistry , Flavonoids/pharmacology , Inflammation Mediators/metabolism , Leukocytes/drug effects , Lipopolysaccharides , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Models, Biological , Oxidative Stress/drug effects , Phosphorylation/drug effects , Pneumonia/blood , Pneumonia/complications , Pneumonia/drug therapy , Pneumonia/physiopathology , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transcription Factor RelA/metabolismABSTRACT
BACKGROUND: Gastric contents aspiration in humans is a risk factor for severe respiratory failure with elevated mortality. Although aspiration-induced local lung inflammation has been studied in animal models, little is known about extrapulmonary effects of aspiration. We investigated whether a single orotracheal instillation of whole gastric fluid elicits a liver acute phase response and if this response contributes to enrich the alveolar spaces with proteins having antiprotease activity. METHODS: In anesthetized Sprague-Dawley rats receiving whole gastric fluid, we studied at different times after instillation (4 h -7 days): changes in blood cytokines and acute phase proteins (fibrinogen and the antiproteases alpha1-antitrypsin and alpha2-macroglobulin) as well as liver mRNA expression of the two antiproteases. The impact of the systemic changes on lung antiprotease defense was evaluated by measuring levels and bioactivity of antiproteases in broncho-alveolar lavage fluid (BALF). Markers of alveolar-capillary barrier derangement were also studied. Non-parametric ANOVA (Kruskall-Wallis) and linear regression analysis were used. RESULTS: Severe peribronchiolar injury involving edema, intra-alveolar proteinaceous debris, hemorrhage and PMNn cell infiltration was seen in the first 24 h and later resolved. Despite a large increase in several lung cytokines, only IL-6 was found elevated in blood, preceding increased liver expression and blood concentration of both antiproteases. These changes, with an acute phase response profile, were significantly larger for alpha2-macroglobulin (40-fold increment in expression with 12-fold elevation in blood protein concentration) than for alpha1-antitrypsin (2-3 fold increment in expression with 0.5-fold elevation in blood protein concentration). Both the increment in capillary-alveolar antiprotease concentration gradient due to increased antiprotease liver synthesis and a timely-associated derangement of the alveolar-capillary barrier induced by aspiration, contributed a 58-fold and a 190-fold increase in BALF alpha1-antitrypsin and alpha2-macroglobulin levels respectively (p < 0.001). CONCLUSIONS: Gastric contents-induced acute lung injury elicits a liver acute phase response characterized by increased mRNA expression of antiproteases and elevation of blood antiprotease concentrations. Hepatic changes act in concert with derangement of the alveolar capillary barrier to enrich alveolar spaces with antiproteases. These findings may have significant implications decreasing protease burden, limiting injury in this and other models of acute lung injury and likely, in recurrent aspiration.
Subject(s)
Acute Lung Injury/enzymology , Acute-Phase Reaction/enzymology , Liver/metabolism , Pregnancy-Associated alpha 2-Macroglobulins/biosynthesis , Pulmonary Alveoli/enzymology , Respiratory Aspiration of Gastric Contents/complications , alpha 1-Antitrypsin/biosynthesis , Acute Lung Injury/blood , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Acute-Phase Reaction/blood , Acute-Phase Reaction/etiology , Acute-Phase Reaction/pathology , Animals , Blood-Air Barrier/enzymology , Blood-Air Barrier/pathology , Disease Models, Animal , Enzyme Induction , Inflammation Mediators/blood , Interleukin-6/blood , Male , Pregnancy-Associated alpha 2-Macroglobulins/genetics , Pulmonary Alveoli/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats, Sprague-Dawley , Time Factors , alpha 1-Antitrypsin/blood , alpha 1-Antitrypsin/geneticsABSTRACT
PURPOSE: To investigate the regulatory roles of neutrophil elastase (NE) and matrix metalloproteinase-9 (MMP-9) in lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. METHODS: To construct LPS-induced ALI mouse models, wild-type C57BL/6 mice were administered 5.0 mg/kg of LPS through endotracheal, and/or 1.0 mg/kg of ONO-5046, and/or 20.0 mg/kg of chemically modified tetracycline-3 (CMT-3) by gavage. The levels of MMP-9, tissue inhibitor of metalloprotease-1, interleukin (IL)-6 were detected by real time RT-PCR at 6 h, 24 h and 48 h, and tumor necrosis factor (TNF), lung wet-dry weight ratio, white blood cell (WBC) count and polymorphonuclear (PMN) count in bronchoalveolar lavage fluid (BALF) were tested at 48 h after administration. The 5-day survival analysis of the ALI mice was also performed. RESULTS: Both ONO-5046 and CMT-3, regardless of being used individually or combined, significantly reduced the levels of MMP-9, IL-6, and TNF in lung tissue as well as in BALF, and the WBC and PMN count in BALF. Combined treatment with ONO-5046 and CMT-3 remarkably improved the survival rate of ALI mice. CONCLUSION: Neutrophil elastase synergizes with matrix metalloproteinase-9 to promote and regulate the release of inflammatory mediators and the infiltration of inflammatory cells, consequently affecting the survival of lipopolysaccharide-induced acute lung injury mice.
Subject(s)
Acute Lung Injury/enzymology , Glycine/analogs & derivatives , Leukocyte Elastase/metabolism , Matrix Metalloproteinase 9/metabolism , Sulfonamides/administration & dosage , Tetracyclines/administration & dosage , Acute Lung Injury/blood , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Glycine/administration & dosage , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Leukocyte Count , Leukocyte Elastase/drug effects , Lipopolysaccharides , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase 9/drug effects , Mice, Inbred C57BL , Neutrophils , Survival Analysis , Time Factors , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tumor Necrosis Factors/metabolismABSTRACT
ABSTRACT PURPOSE: To investigate the regulatory roles of neutrophil elastase (NE) and matrix metalloproteinase-9 (MMP-9) in lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. METHODS: To construct LPS-induced ALI mouse models, wild-type C57BL/6 mice were administered 5.0 mg/kg of LPS through endotracheal, and/or 1.0 mg/kg of ONO-5046, and/or 20.0 mg/kg of chemically modified tetracycline-3 (CMT-3) by gavage. The levels of MMP-9, tissue inhibitor of metalloprotease-1, interleukin (IL)-6 were detected by real time RT-PCR at 6 h, 24 h and 48 h, and tumor necrosis factor (TNF), lung wet-dry weight ratio, white blood cell (WBC) count and polymorphonuclear (PMN) count in bronchoalveolar lavage fluid (BALF) were tested at 48 h after administration. The 5-day survival analysis of the ALI mice was also performed. RESULTS: Both ONO-5046 and CMT-3, regardless of being used individually or combined, significantly reduced the levels of MMP-9, IL-6, and TNF in lung tissue as well as in BALF, and the WBC and PMN count in BALF. Combined treatment with ONO-5046 and CMT-3 remarkably improved the survival rate of ALI mice. CONCLUSION: Neutrophil elastase synergizes with matrix metalloproteinase-9 to promote and regulate the release of inflammatory mediators and the infiltration of inflammatory cells, consequently affecting the survival of lipopolysaccharide-induced acute lung injury mice.
Subject(s)
Animals , Sulfonamides/administration & dosage , Tetracyclines/administration & dosage , Leukocyte Elastase/metabolism , Matrix Metalloproteinase 9/metabolism , Acute Lung Injury/enzymology , Glycine/analogs & derivatives , Time Factors , Bronchoalveolar Lavage Fluid/cytology , Survival Analysis , Lipopolysaccharides , Interleukin-6/metabolism , Inflammation Mediators/metabolism , Leukocyte Elastase/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase 9/drug effects , Tumor Necrosis Factors/metabolism , Disease Models, Animal , Acute Lung Injury/chemically induced , Acute Lung Injury/blood , Glycine/administration & dosage , Leukocyte Count , Mice, Inbred C57BL , NeutrophilsABSTRACT
BACKGROUND: The radiation-induced lung injury is a common complication from radiotherapy in lung cancer. CpG ODN is TLR9 activator with potential immune modulatory effects and sensitization of radiotherapy in lung cancer. This study aimed to examine the effect of CpG ODN on acute radiation-induced lung injury in mice. METHODS AND RESULTS: The mouse model of radiation-induced lung injury was established by a single dose of 20 Gy X-rays exposure to the left lung. The results showed that the pneumonia score was lower in RT+CpG group than in RT group on 15th and 30th days. Compared with RT group, CpG ODN reduced the serum concentrations of MDA (P < 0.05) and increased the serum concentrations of SOD, GSH (P < 0.05). The serum concentration of TNF-α in RT+CpG group was lower on 15th and 30th days post-irradiation (P < 0.05). CONCLUSION: The study demonstrated that CpG ODN has preventive effects of acute radiation-induced lung injury in mice. Lung inflammatory reaction and oxidative stress are promoted in the initiation of radiation-induced pneumonia. CpG ODN may reduce the injury of reactive oxygen species and adjust the serum TNF-α concentration in the mice after irradiation, which reduces the generation of the inflammatory cytokines.
Subject(s)
Acute Lung Injury/prevention & control , Oligodeoxyribonucleotides/pharmacology , Radiation Injuries, Experimental/prevention & control , Acute Lung Injury/blood , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Glutathione/blood , Malondialdehyde/blood , Mice , Pneumonia/etiology , Pneumonia/pathology , Pneumonia/prevention & control , Radiation Injuries, Experimental/blood , Reproducibility of Results , Severity of Illness Index , Superoxide Dismutase/blood , Time Factors , Tumor Necrosis Factor-alpha/bloodABSTRACT
INTRODUCTION: The benefits of prone position ventilation are well demonstrated in the severe forms of acute respiratory distress syndrome, but not in the milder forms. We investigated the effects of prone position on arterial blood gases, lung inflammation, and histology in an experimental mild acute lung injury (ALI) model. METHODS: ALI was induced in Wistar rats by intraperitoneal Escherichia coli lipopolysaccharide (LPS, 5 mg/kg). After 24 h, the animals with PaO2/FIO2 between 200 and 300 mmHg were randomized into 2 groups: prone position (n = 6) and supine position (n = 6). Both groups were compared with a control group (n = 5) that was ventilated in the supine position. All of the groups were ventilated for 1 h with volume-controlled ventilation mode (tidal volume = 6 ml/kg, respiratory rate = 80 breaths/min, positive end-expiratory pressure = 5 cmH2O, inspired oxygen fraction = 1) RESULTS: Significantly higher lung injury scores were observed in the LPS-supine group compared to the LPS-prone and control groups (0.32 ± 0.03; 0.17 ± 0.03 and 0.13 ± 0.04, respectively) (p < 0.001), mainly due to a higher neutrophil infiltration level in the interstitial space and more proteinaceous debris that filled the airspaces. Similar differences were observed when the gravity-dependent lung regions and non-dependent lung regions were analyzed separately (p < 0.05). The BAL neutrophil content was also higher in the LPS-supine group compared to the LPS-prone and control groups (p < 0.05). There were no significant differences in the wet/dry ratio and gas exchange levels. CONCLUSIONS: In this experimental extrapulmonary mild ALI model, prone position ventilation for 1 h, when compared with supine position ventilation, was associated with lower lung inflammation and injury.
Subject(s)
Acute Lung Injury/prevention & control , Lipopolysaccharides , Lung/pathology , Pneumonia/prevention & control , Positive-Pressure Respiration/methods , Prone Position , Ventilator-Induced Lung Injury/prevention & control , Acute Lung Injury/blood , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Injections, Intraperitoneal , Lung/physiopathology , Male , Neutrophil Infiltration , Pneumonia/blood , Pneumonia/chemically induced , Pneumonia/pathology , Positive-Pressure Respiration/adverse effects , Rats, Wistar , Respiratory Rate , Supine Position , Tidal Volume , Ventilator-Induced Lung Injury/blood , Ventilator-Induced Lung Injury/chemically induced , Ventilator-Induced Lung Injury/pathologyABSTRACT
BACKGROUND: The radiation-induced lung injury is a common complication from radiotherapy in lung cancer. CpG ODN is TLR9 activator with potential immune modulatory effects and sensitization of radiotherapy in lung cancer. This study aimed to examine the effect of CpG ODN on acute radiation-induced lung injury in mice. METHODS AND RESULTS: The mouse model of radiation-induced lung injury was established by a single dose of 20 Gy X-rays exposure to the left lung. The results showed that the pneumonia score was lower in RT+CpG group than in RT group on 15th and 30th days. Compared with RT group, CpG ODN reduced the serum concentrations of MDA (P < 0.05) and increased the serum concentrations of SOD, GSH (P < 0.05). The serum concentration of TNF-α in RT+CpG group was lower on 15th and 30th days post-irradiation (P < 0.05). CONCLUSION: The study demonstrated that CpG ODN has preventive effects of acute radiation-induced lung injury in mice. Lung inflammatory reaction and oxidative stress are promoted in the initiation of radiation-induced pneumonia. CpG ODN may reduce the injury of reactive oxygen species and adjust the serum TNF-α concentration in the mice after irradiation, which reduces the generation of the inflammatory cytokines.
Subject(s)
Animals , Mice , Oligodeoxyribonucleotides/pharmacology , Radiation Injuries, Experimental/prevention & control , Acute Lung Injury/prevention & control , Pneumonia/etiology , Pneumonia/pathology , Pneumonia/prevention & control , Radiation Injuries, Experimental/blood , Superoxide Dismutase/blood , Time Factors , Severity of Illness Index , Enzyme-Linked Immunosorbent Assay , Reproducibility of Results , Tumor Necrosis Factor-alpha/blood , Disease Models, Animal , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Acute Lung Injury/blood , Glutathione/blood , Malondialdehyde/bloodABSTRACT
Acute lung injury (ALI) is a common complication associated with septic shock that directly influences the prognosis of sepsis patients. Currently, one of the main supportive treatment modalities for septic shock is fluid resuscitation. The use of hypertonic saline (HS: 7.5% NaCl) for fluid resuscitation has been described as a promising therapy in experimental models of sepsis-induced ALI, but it has failed to produce similar results in clinical practice. Thus, we compared experimental timing versus clinical timing effectiveness (i.e., early vs. late fluid resuscitation) after the inflammatory scenario was established in a rat model of bacterial lipopolysaccharide-induced ALI. We found that late fluid resuscitation with hypertonic saline (NaCl 7.5%) did not reduce the mortality rates of animals compared with the mortality late associated with early treatment. Late fluid resuscitation with both hypertonic and normal saline increased pulmonary inflammation, decreased pulmonary function, and induced pulmonary injury by elevating metalloproteinase-2 and metalloproteinase-9 activity and collagen deposition in the animals, unlike early treatment. The animals with lipopolysaccharide-induced ALI that received late resuscitation with any kind of fluids demonstrated aggravated pulmonary injury and respiratory function. Moreover, we showed that the therapeutic window for a beneficial effect of fluid resuscitation with hypertonic saline is very narrow.
Subject(s)
Acute Lung Injury/blood , Acute Lung Injury/chemically induced , Lipopolysaccharides/adverse effects , Resuscitation/methods , Saline Solution, Hypertonic/therapeutic use , Animals , Collagen/chemistry , Cytokines/metabolism , Disease Models, Animal , Endotoxins/chemistry , Fluid Therapy/methods , Heat-Shock Proteins/metabolism , Inflammation , Lung/physiopathology , Lung Injury/physiopathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Prognosis , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Sepsis/physiopathology , Shock, Septic/physiopathology , Sodium Chloride/chemistry , Time FactorsABSTRACT
OBJECTIVE: To evaluate the effect of the nasal administration of live and heat-killed Lactobacillus rhamnosus CRL1505 (Lr1505) on immune-coagulative response during influenza virus (IFV) infection to improve survival and reduce lung injury. METHODS: Six-week-old BALB/c mice were treated with live or heat-killed Lr1505 by the nasal route during two consecutive days. Treated and untreated control mice were then nasally challenged with IFV. RESULTS: Both viable and non-viable Lr1505 protected infected mice by reducing pulmonary injury and lung viral loads trough several mechanisms: (a) Inflammatory cytokines were efficiently regulated allowing higher clearance of virus and reduction of inflammatory lung tissue damage, associated to higher levels of the regulatory cytokine IL-10. (b) The antiviral immune response was enhanced with improved levels of type I interferons, CD4(+)IFN-γ(+) lymphocytes, and lung CD11c(+)CD11b(low)CD103(+) and CD11c(+)CD11b(high)CD103(-) dendritic cells. (c) The procoagulant state was reversed mainly by down-regulating tissue factor expression and restoring thrombomodulin levels in lung. The capacity of Lr1505 to improve the outcome of IFV infection would be related to its ability to beneficially modulate lung TLR3-triggered immune response. CONCLUSIONS: Our work is the first to demonstrate the ability of an immunobiotic strain to beneficially modulate inflammation-coagulation interactions during IFV infection. Interestingly, non-viable L. rhamnosus CRL1505 was as effective as the viable strain to beneficially modulate respiratory antiviral immune response.
Subject(s)
Acute Lung Injury/immunology , Immunologic Factors/pharmacology , Lacticaseibacillus rhamnosus , Orthomyxoviridae Infections/immunology , Probiotics/pharmacology , Acute Lung Injury/blood , Acute Lung Injury/pathology , Acute Lung Injury/virology , Administration, Intranasal , Animals , Blood Coagulation , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/blood , Cytokines/immunology , Leukocyte Count , Lung/immunology , Lung/pathology , Lung/virology , Male , Mice, Inbred BALB C , Orthomyxoviridae , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Platelet Count , Poly I-C/pharmacologyABSTRACT
The aim of this study was to determine the effects of previous administration of metyrapone (met) on the acute lung injury (ALI) induced by caecal ligation and puncture (CLP) and to explore met's relationship with endogenous glucocorticoids (GCs) as measured by inflammatory, oxidative and functional parameters. One hundred and thirty-five Wistar rats were divided into three main groups: Control (Naïve), Sham and CLP. The animals received pretreatment one hour before surgery. The Naïve group did not undergo any procedure or pretreatment. The Sham group only had the caecum exposed and was pretreated with saline. The CLP group was divided into three pretreatments: metyrapone (CLP met 50 mg/kg i.p.), dexamethasone (CLP dex 0.5 mg/kg i.p.) or saline (CLP sal equivalent volume of 0.9% NaCl). Analyses were performed after 6 and 24 h of sepsis. Previous administration of met significantly increased inflammatory cells, as well as myeloperoxidase (MPO) activity in the lung tissue and alveolar collapsed area, with consequent impairment of respiratory mechanics being observed compared to Sham and Naïve; CLP sal exhibited similar results to those of met. The met reduced corticosterone (CCT) levels and dramatically increased hydrogen peroxide (H2 O2 ) levels in the lung tissue compared to CLP sal. Our results suggest that previous administration of met may have contributed to increased pulmonary oxidative stress and increased mortality by mechanisms dependent of endogenous GC.
Subject(s)
Acute Lung Injury/chemically induced , Corticosterone/biosynthesis , Enzyme Inhibitors/toxicity , Lung/drug effects , Metyrapone/toxicity , Shock, Septic/complications , Steroid 11-beta-Hydroxylase/antagonists & inhibitors , Acute Lung Injury/blood , Acute Lung Injury/pathology , Acute Lung Injury/physiopathology , Animals , Corticosterone/blood , Disease Models, Animal , Down-Regulation , Hydrogen Peroxide/metabolism , Inflammation Mediators/metabolism , Lung/metabolism , Lung/pathology , Lung/physiopathology , Male , Oxidative Stress/drug effects , Rats, Wistar , Respiratory Mechanics/drug effects , Steroid 11-beta-Hydroxylase/metabolism , Time FactorsABSTRACT
BACKGROUND: Cardiac output (CO) measurement is not a standard of care for critically ill children, but it can be estimated by indirect methods such as veno-arterial pCO2 difference (ΔVACO2). AIM: To determine the correlation between CO and ΔVACO2 and evaluate the usefulness of ΔVACO2 in the diagnosis of low CO in an experimental pediatric model. MATERIALS AND METHODS: Thirty piglets weighing 4.8 ± 0.35 kg were anesthetized and monitored with transpulmonary thermodilution. Lung injury was induced with tracheal instillation of Tween 20®. Serial measurements of central venous and arterial blood gases, as well as CO, were obtained at baseline, 1, 2 and 4 h after lung injury induction. Low cardiac output (LCO) was defined as CO lower than 2.5 Llminlm². RESULTS: There was an inverse correlation between CO and ΔVACO2 (r = -0.36, p < 0.01). ΔVACO2 was 14 ± 8 mmHg in LCO state and 8 ± 6 mmHg when this condition was not present (p < 0.01). Area under the receiver operating characteristic (ROC) curves of ΔVACO2 and LCO state was 0.78 (0.68-0.86). The best cut-point was 8.9 mmHg to determine LCO with a sensibility 0.78, specificity 0.7, positive predictive value 0.27 and negative predictive value 0.96. CONCLUSIONS: In this model there was an inverse correlation between ΔVACO2 and CO. The best cutoff value to discard LCO was ΔVACO2 of 8.9 mmHg, indicating that under this value the presence of LCO is very unlikely.
Subject(s)
Acute Lung Injury/blood , Carbon Dioxide/blood , Cardiac Output, Low/blood , Animals , Area Under Curve , Blood Gas Analysis , Cardiac Output, Low/diagnosis , Disease Models, Animal , Predictive Value of Tests , Swine , ThermodilutionABSTRACT
UNLABELLED: Acetaminophen (APAP) is a safe analgesic and antipyretic drug. However, APAP overdose leads to massive hepatocyte death. Cell death during APAP toxicity occurs by oncotic necrosis, in which the release of intracellular contents can elicit a reactive inflammatory response. We have previously demonstrated that an intravascular gradient of chemokines and mitochondria-derived formyl peptides collaborate to guide neutrophils to sites of liver necrosis by CXC chemokine receptor 2 (CXCR2) and formyl peptide receptor 1 (FPR1), respectively. Here, we investigated the role of CXCR2 chemokines and mitochondrial products during APAP-induced liver injury and in liver neutrophil influx and hepatotoxicity. During APAP overdose, neutrophils accumulated into the liver, and blockage of neutrophil infiltration by anti-granulocyte receptor 1 depletion or combined CXCR2-FPR1 antagonism significantly prevented hepatotoxicity. In agreement with our in vivo data, isolated human neutrophils were cytotoxic to HepG2 cells when cocultured, and the mechanism of neutrophil killing was dependent on direct contact with HepG2 cells and the CXCR2-FPR1-signaling pathway. Also, in mice and humans, serum levels of both mitochondrial DNA (mitDNA) and CXCR2 chemokines were higher during acute liver injury, suggesting that necrosis products may reach remote organs through the circulation, leading to a systemic inflammatory response. Accordingly, APAP-treated mice exhibited marked systemic inflammation and lung injury, which was prevented by CXCR2-FPR1 blockage and Toll-like receptor 9 (TLR9) absence (TLR9(-/-) mice). CONCLUSION: Chemokines and mitochondrial products (e.g., formyl peptides and mitDNA) collaborate in neutrophil-mediated injury and systemic inflammation during acute liver failure. Hepatocyte death is amplified by liver neutrophil infiltration, and the release of necrotic products into the circulation may trigger a systemic inflammatory response and remote lung injury.
Subject(s)
Acute-Phase Reaction/metabolism , Chemokines/metabolism , DNA, Mitochondrial/blood , Liver Failure, Acute/immunology , Liver/pathology , Neutrophils/immunology , Receptors, Formyl Peptide/metabolism , Acetaminophen , Acute Lung Injury/blood , Acute Lung Injury/immunology , Acute-Phase Reaction/immunology , Adolescent , Adult , Analysis of Variance , Animals , Cell Movement , Chemokines/blood , Chemokines/immunology , Child , Coculture Techniques , Female , Hep G2 Cells , Humans , Interleukin-8/blood , Liver/metabolism , Liver Failure, Acute/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitochondrial Proteins/immunology , Mitochondrial Proteins/metabolism , Necrosis/immunology , Receptors, Formyl Peptide/immunology , Receptors, Interleukin-8B/blood , Receptors, Interleukin-8B/immunology , Receptors, Interleukin-8B/metabolism , Signal Transduction , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Young AdultABSTRACT
Background: Cardiac output (CO) measurement is not a standard of care for critically ill children, but it can be estimated by indirect methods such as veno-arterial pCO2 difference (ΔVACO2). Aim: To determine the correlation between CO and ΔVACO2 and evaluate the usefulness of ΔVACO2 in the diagnosis of low CO in an experimental pediatric model. Materials and Methods: Thirty piglets weighing 4.8 ± 0.35 kg were anesthetized and monitored with transpulmonary thermodilution. Lung injury was induced with tracheal instillation of Tween 20®. Serial measurements of central venous and arterial blood gases, as well as CO, were obtained at baseline, 1, 2 and 4 h after lung injury induction. Low cardiac output (LCO) was defined as CO lower than 2.5 Llminlm². Results: There was an inverse correlation between CO and ΔVACO2 (r = -0.36, p < 0.01). ΔVACO2 was 14 ± 8 mmHg in LCO state and 8 ± 6 mmHg when this condition was not present (p < 0.01). Area under the receiver operating characteristic (ROC) curves of ΔVACO2 and LCO state was 0.78 (0.68-0.86). The best cut-point was 8.9 mmHg to determine LCO with a sensibility 0.78, specificity 0.7, positive predictive value 0.27 and negative predictive value 0.96. Conclusions: In this model there was an inverse correlation between ΔVACO2 and CO. The best cutoff value to discard LCO was ΔVACO2 of 8.9 mmHg, indicating that under this value the presence of LCO is very unlikely.
Subject(s)
Animals , Acute Lung Injury/blood , Carbon Dioxide/blood , Cardiac Output, Low/blood , Area Under Curve , Blood Gas Analysis , Cardiac Output, Low/diagnosis , Disease Models, Animal , Predictive Value of Tests , Swine , ThermodilutionABSTRACT
We tested the hypothesis that at the early phase of acute lung injury (ALI) the degree of endothelium injury may predict lung parenchyma remodelling. For this purpose, two models of extrapulmonary ALI induced by Escherichia coli lipopolysaccharide (ALI-LPS) or cecal ligation and puncture (ALI-CLP) were developed in mice. At day 1, these models had similar degrees of lung mechanical compromise, epithelial damage, and intraperitoneal inflammation, but endothelial lesion was greater in ALI-CLP. A time course analysis revealed, at day 7: ALI-CLP had higher degrees of epithelial lesion, denudation of basement membrane, endothelial damage, elastic and collagen fibre content, neutrophils in bronchoalveolar lavage fluid (BALF), peritoneal fluid and blood, levels of interleukin-6, KC (murine analogue of IL-8), and transforming growth factor-ß in BALF. Conversely, the number of lung apoptotic cells was similar in both groups. In conclusion, the intensity of fibroelastogenesis was affected by endothelium injury in addition to the maintenance of epithelial damage and intraperitoneal inflammation.
Subject(s)
Acute Lung Injury/pathology , Endothelium/injuries , Endothelium/physiopathology , Fibroblasts/physiology , Lung/pathology , Acute Lung Injury/blood , Acute Lung Injury/classification , Acute Lung Injury/etiology , Analysis of Variance , Animals , Antigens, CD34/metabolism , Bronchoalveolar Lavage Fluid , Cecal Diseases/complications , Collagen/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Fibroblasts/ultrastructure , Lipopolysaccharides , Lung/physiopathology , Lung/ultrastructure , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission/methods , Respiratory Mechanics/drug effects , Respiratory Mechanics/physiology , Time Factors , Transcription FactorsABSTRACT
Shock and resuscitation render patients more susceptible to acute lung injury due to an exacerbated immune response to subsequent inflammatory stimuli. To study the role of innate immunity in this situation, we investigated acute lung injury in an experimental model of ischemia-reperfusion (I-R) followed by an early challenge with live bacteria. Conscious rats (N = 8 in each group) were submitted to controlled hemorrhage and resuscitated with isotonic saline (SS, 0.9 percent NaCl) or hypertonic saline (HS, 7.5 percent NaCl) solution, followed by intratracheal or intraperitoneal inoculation of Escherichia coli. After infection, toll-like receptor (TLR) 2 and 4 mRNA expression was monitored by RT-PCR in infected tissues. Plasma levels of tumor necrosis factor α and interleukins 6 and 10 were determined by ELISA. All animals showed similar hemodynamic variables, with mean arterial pressure decreasing to nearly 40 mmHg after bleeding. HS or SS used as resuscitation fluid yielded equal hemodynamic results. Intratracheal E. coli inoculation per se induced a marked neutrophil infiltration in septa and inside the alveoli, while intraperitoneal inoculation-associated neutrophils and edema were restricted to the interseptal space. Previous I-R enhanced lung neutrophil infiltration upon bacterial challenge when SS was used as reperfusion fluid, whereas neutrophil influx was unchanged in HS-treated animals. No difference in TLR expression or cytokine secretion was detected between groups receiving HS or SS. We conclude that HS is effective in reducing the early inflammatory response to infection after I-R, and that this phenomenon is achieved by modulation of factors other than expression of innate immunity components.