Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 317
Filter
1.
AAPS J ; 26(3): 47, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622374

ABSTRACT

BACKGROUND: Sepsis-induced acute lung injury (ALI) is one of the serious life-threatening complications of sepsis and is pathologically associated with mitochondrial dysfunction. Ginsenoside Rg1 has good therapeutic effects on ALI. Herein, the pharmacological effects of Rg1 in sepsis-induced ALI were investigated. METHODS: Sepsis-induced ALI models were established by CLP operation and LPS treatment. HE staining was adopted to analyze lung pathological changes. The expression and secretion of cytokines were measured by RT-qPCR and ELISA. Cell viability and apoptosis were assessed by MTT assay, flow cytometry and TUNEL staining. ROS level and mitochondrial membrane potential (MMP) were analyzed using DHE probe and JC-1 staining, respectively. FBXO3 m6A level was assessed using MeRIP assay. The interactions between FBXO3, YTHDF1, and PGC-1α were analyzed by Co-IP or RIP. RESULTS: Rg1 administration ameliorated LPS-induced epithelial cell inflammation, apoptosis, and mitochondrial dysfunction in a dose-dependent manner. Mechanically, Rg1 reduced PGC-1α ubiquitination modification level by inhibiting FBXO3 expression m6A-YTHDF1 dependently. As expected, Rg1's mitigative effect on LPS-induced inflammation, apoptosis and mitochondrial dysfunction in lung epithelial cells was abolished by FBXO3 overexpression. Moreover, FBXO3 upregulation eliminated the restoring effect of Rg1 on CLP-induced lung injury in rats. CONCLUSION: Rg1 activated PGC-1α/Nrf2 signaling pathway by reducing FBXO3 stability in an m6A-YTHDF1-dependent manner to improve mitochondrial function in lung epithelial cells during sepsis-induced ALI progression.


Subject(s)
Acute Lung Injury , Ginsenosides , Mitochondrial Diseases , Sepsis , Rats , Animals , NF-E2-Related Factor 2/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/therapeutic use , Signal Transduction , Acute Lung Injury/etiology , Acute Lung Injury/complications , Inflammation , Sepsis/complications , Sepsis/drug therapy , Mitochondrial Diseases/complications
2.
Photobiomodul Photomed Laser Surg ; 42(2): 148-158, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38301209

ABSTRACT

Background: Sepsis-induced acute lung injury (ALI) is a clinical syndrome characterized by excessive uncontrolled inflammation. Photobiomodulation such as light-emitting diode (LED) irradiation has been used to attenuate inflammatory disease. Objective: The protective effect of 630 nm LED irradiation on sepsis-induced ALI remains unknown. The purpose of this study was to investigate the role of 630 nm LED irradiation in sepsis-induced ALI and its underlying mechanism. Methods and results: C57BL/6 mice were performed cecal ligation and puncture (CLP) for 12 h to generate experimental sepsis models. Histopathology analysis showed that alveolar injury, inflammatory cells infiltration, and hemorrhage were suppressed in CLP mice after 630 nm LED irradiation. The ratio of wet/dry weigh of lung tissue was significantly inhibited by irradiation. The number of leukocytes was reduced in bronchoalveolar lavage fluid. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) results and enzyme-linked immunosorbent assay showed that 630 nm LED irradiation significantly inhibited the mRNA and protein levels of M1 macrophage-related genes in the lung of CLP-induced septic mice. Meanwhile, LED irradiation significantly inhibited signal transducer and activator of transcription 1 (STAT1) phosphorylation in the lung of septic mice. In vitro experiments showed that 630 nm LED irradiation significantly inhibited M1 genes mRNA and protein expression in THP-1-derived M1 macrophages without affecting the cell viability. LED irradiation also significantly inhibited the level of STAT1 phosphorylation in THP-1-derived M1 macrophages. Conclusions: We concluded that 630 nm LED is promising as a treatment against ALI through inhibiting M1 macrophage polarization, which is associated with the downregulation of STAT1 phosphorylation.


Subject(s)
Acute Lung Injury , Low-Level Light Therapy , Sepsis , Mice , Animals , Mice, Inbred C57BL , Acute Lung Injury/complications , Acute Lung Injury/drug therapy , Macrophages , Sepsis/complications , Sepsis/radiotherapy , Sepsis/drug therapy , RNA, Messenger
3.
Eur J Pharmacol ; 963: 176219, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38040079

ABSTRACT

Sepsis-associated lung injury often coexists with intestinal dysfunction. Butyrate, an essential gut microbiota metabolite, participates in gut-lung crosstalk and has immunoregulatory effects. This study aims to investigate the effect and mechanism of sodium butyrate (NaB) on lung injury. Sepsis-associated lung injury was established in mice by cecal ligation and puncture (CLP). Mice in treatment groups received NaB gavage after surgery. The survival rate, the oxygenation index and the lung wet-to-dry weight (W/D) ratio were calculated respectively. Pulmonary and intestinal histologic changes were observed. The total protein concentration in bronchoalveolar lavage fluid (BALF) was measured, and inflammatory factors in serum and BALF were examined. Diamine oxidase (DAO), lipopolysaccharide (LPS), and surfactant-associated protein D (SP-D) levels in serum and amphiregulin in lung tissue were assessed. Intercellular junction protein expression in the lung and intestinal tissues were examined. Changes in immune cells were analyzed. NaB treatment improved the survival rate, the oxygenation index and the histologic changes. NaB decreased the W/D ratio, total protein concentration, and the levels of proinflammatory cytokines, as well as SP-D, DAO and LPS, while increased the levels of anti-inflammatory cytokines and amphiregulin. The intercellular junction protein expression were improved by NaB. Furthermore, the CD4+/CD8+ T-cell ratio and the proportion of CD4+Foxp3+ regulatory T cells (Tregs) were increased by NaB. Our data suggested that NaB gavage effectively improved the survival rate and mitigated lung injury in CLP mice. The possible mechanism was that NaB augmented CD4+Foxp3+ Tregs and enhanced the barrier function of the gut and the lung.


Subject(s)
Acute Lung Injury , Sepsis , Mice , Animals , Acute Lung Injury/etiology , Acute Lung Injury/complications , Butyric Acid/pharmacology , Butyric Acid/therapeutic use , Butyric Acid/metabolism , Amphiregulin/metabolism , T-Lymphocytes, Regulatory/metabolism , Lipopolysaccharides/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Lung/pathology , Cytokines/metabolism , Transcription Factors/metabolism , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , Forkhead Transcription Factors/metabolism
4.
BMJ Open Respir Res ; 10(1)2023 12 14.
Article in English | MEDLINE | ID: mdl-38097355

ABSTRACT

BACKGROUND: Supraphysiological oxygen administration causes unfavourable clinical outcomes in various diseases. This study aimed to determine whether hyperoxia would be associated with increased mortality in patients with severe infection. METHODS: A post-hoc analysis of a nationwide multicentre prospective observational study on sepsis (SPICE Study) was conducted, including adult patients admitted to the intensive care unit with available arterial partial pressure of oxygen (PaO2) at the treatment initiation for severe infection. Hyperoxia was defined as a PaO2 level of ≥300 mm Hg and in-hospital mortality was compared between patients with and without hyperoxia. RESULTS: Of the 563 patients eligible for the study, 49 had hyperoxia at treatment initiation for severe infection. The in-hospital all-cause mortality rates of patients with and without hyperoxia were 14 (29.2%) and 90 (17.6%), respectively. Inverse probability weighting analyses with propensity scores revealed the association between hyperoxia and increased in-hospital mortality rate (28.8% vs 18.8%; adjusted OR 1.75 (1.03 to 2.97); p=0.038), adjusting for patient demographics, comorbidities, site of infection, severity of infection, haemodynamic and respiratory status, laboratory data and location of patient at infection development. Acute lung injury developed more frequently in patients with hyperoxia on the following days after infection treatment, whereas sepsis-related mortality was comparable regardless of hyperoxia exposure. CONCLUSION: Hyperoxia with PaO2 ≥300 mm Hg at treatment initiation of severe infection was associated with an increased in-hospital mortality rate in patients requiring intensive care. The amount of oxygen to administer to patients with severe infection should be carefully determined. TRIAL REGISTRATION NUMBER: University Hospital Medical Information Network Clinical Trial Registry (UMIN000027452).


Subject(s)
Acute Lung Injury , Hyperoxia , Sepsis , Adult , Humans , Hyperoxia/complications , Retrospective Studies , Oxygen , Acute Lung Injury/complications
5.
Int Immunopharmacol ; 124(Pt B): 111063, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37857120

ABSTRACT

Sepsis-induced lung injury is an acute hypoxic respiratory insufficiency caused by systemic infectious factors that results in alveolar epithelial cell and capillary endothelial cell injury, diffuse pulmonary interstitial edema, and alveolar edema. Heme oxygenase (HO)-1 is usually associated with inflammation and has anti-inflammatory effects. Autophagy is a degradation pathway that eliminates cellular metabolic waste and plays an important protective role during stress. The phosphatidylinositol 3-kinase/ protein kinase B (PI3K/Akt) signaling pathway plays a key role in mediating cellular responses to inflammatory reactions. Therefore, we hypothesized that HO-1 is associated with autophagy and regulated by the PI3K/Akt signaling pathway in mice with sepsis-induced lung injury. Sepsis-induced lung injury was induced in mice using cecal ligation and puncture (CLP). Hemin or Sn-protoporphyrin IX (SnPP) was administered via intraperitoneal injection before surgery. Survival rates were observed during days 1-7 after the surgery; lung histology was discerned 24 h after the surgery; pro-inflammatory and anti-inflammatory factors in plasma and lung tissue were measured using enzyme-linked immunosorbent assay (ELISA); HO-1, Beclin-1, microtubule-associated protein 1 light chain 3B (LC3B)-II, p62 and lysosome associated membrane protein (LAMP)2 protein expression levels were measured 24 h after the surgery; HO-1 and LC3B-II protein expression levels were observed using immunofluorescence 24 h after the surgery; and autophagosomes were detected using electron microscopy 24 h after the surgery. Furthermore, when PI3K inhibitors LY294002, PI3K activators Recilisib and hemin were administered before the surgery, Akt, p-Akt, HO-1, and LC3-II levels were measured 24 h post-surgery. We found that HO-1 overexpression increased the survival rate and inhibited sepsis-induced lung injury. HO-1 overexpression attenuated the levels of proinflammatory cytokines (TNF-α, IL-1ß) and increased the anti-inflammatory cytokine (IL-10, HO-1) overexpression. Moreover, HO-1 overexpression was also associated with increased expression of Beclin-1, LC3B-II and LAMP2 protein expression; decreased p62 protein expression; and significantly increased autophagosome formation. The results for HO-1-downregulated mice contrasted with those mentioned above. LY294002 inhibited p-Akt/Akt, HO-1, and LC3B-II protein expression; and hemin reversed the inhibitory effect of LY294002. The protective effect of HO-1 was involved in the mediation of autophagy, which may be regulated by the PI3K/Akt signaling pathway during sepsis-induced lung injury in mice.


Subject(s)
Acute Lung Injury , Sepsis , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Beclin-1/metabolism , Hemin/pharmacology , Acute Lung Injury/complications , Cytokines/metabolism , Autophagy , Sepsis/drug therapy , Anti-Inflammatory Agents/therapeutic use , Edema , Heme Oxygenase-1/metabolism
6.
Biomed Pharmacother ; 168: 115719, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37839108

ABSTRACT

Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection and is characterized by multiple biological and clinical features. N6-methyladenosine (m6A) modification is the most common type of RNA modifications in eukaryotes and plays an important regulatory role in various biological processes. Recently, m6A modification has been found to be involved in the regulation of immune responses in sepsis. In addition, several studies have shown that m6A modification is involved in sepsis-induced multiple organ dysfunctions, including cardiovascular dysfunction, acute lung injury (ALI), acute kidney injury (AKI) and etc. Considering the complex pathogenesis of sepsis and the lack of specific therapeutic drugs, m6A modification may be the important bond in the pathophysiological process of sepsis and even therapeutic targets. This review systematically highlights the recent advances regarding the roles of m6A modification in sepsis and sheds light on their use as treatment targets for sepsis.


Subject(s)
Acute Kidney Injury , Acute Lung Injury , Sepsis , Humans , Acute Kidney Injury/drug therapy , Acute Lung Injury/drug therapy , Acute Lung Injury/complications , Sepsis/genetics
7.
Int J Mol Sci ; 24(15)2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37569514

ABSTRACT

Acute lung injury/acute respiratory distress syndrome (ALI/ARDS), triggered by various pathogenic factors inside and outside the lungs, leads to diffuse lung injury and can result in respiratory failure and death, which are typical clinical critical emergencies. Severe acute pancreatitis (SAP), which has a poor clinical prognosis, is one of the most common diseases that induces ARDS. When SAP causes the body to produce a storm of inflammatory factors and even causes sepsis, clinicians will face a two-way choice between anti-inflammatory and anti-infection objectives while considering the damaged intestinal barrier and respiratory failure, which undoubtedly increases the difficulty of the diagnosis and treatment of SAP-ALI/ARDS. For a long time, many studies have been devoted to applying glucocorticoids (GCs) to control the inflammatory response and prevent and treat sepsis and ALI/ARDS. However, the specific mechanism is not precise, the clinical efficacy is uneven, and the corresponding side effects are endless. This review discusses the mechanism of action, current clinical application status, effectiveness assessment, and side effects of GCs in the treatment of ALI/ARDS (especially the subtype caused by SAP).


Subject(s)
Acute Lung Injury , Pancreatitis , Respiratory Distress Syndrome , Respiratory Insufficiency , Sepsis , Humans , Glucocorticoids/therapeutic use , Acute Disease , Pancreatitis/complications , Respiratory Distress Syndrome/pathology , Acute Lung Injury/etiology , Acute Lung Injury/complications , Sepsis/complications
8.
Int Immunopharmacol ; 123: 110638, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37494838

ABSTRACT

INTRODUCTION: Treatment of ARDS caused by smoke inhalation is challenging with no specific therapies available. The aim of this study was to test the efficacy of nebulized adipose-derived mesenchymal stem cells (ASCs) in a well-characterized, clinically relevant ovine model of smoke inhalation injury. MATERIAL AND METHODS: Fourteen female Merino sheep were surgically instrumented 5-7 days prior to study. After induction of acute lung injury (ALI) by cooled cotton smoke insufflation into the lungs (under anesthesia and analgesia), sheep were placed on a mechanical ventilator for 48 hrs and monitored for cardiopulmonary hemodynamics in a conscious state. ASCs were isolated from ovine adipose tissue. Sheep were randomly allocated to two groups after smoke injury: 1) ASCs group (n = 6): 10 million ASCs were nebulized into the airway at 1 hr post-injury; and 2) Control group (n = 8): Nebulized with saline into the airways at 1 hr post-injury. ASCs were labeled with green fluorescent protein (GFP) to trace cells within the lung. ASCs viability was determined in bronchoalveolar lavage fluid (BALF). RESULTS: PaO2/FiO2 in the ASCs group was significantly higher than in the control group (p = 0.001) at 24 hrs. Oxygenation index: (mean airway pressure × FiO2/PaO2) was significantly lower in the ASCs group at 36 hr (p = 0.003). Pulmonary shunt fraction tended to be lower in the ASCs group as compared to the control group. GFP-labelled ASCs were found on the surface of trachea epithelium 48 hrs after injury. The viability of ASCs in BALF was significantly lower than those exposed to the control vehicle solution. CONCLUSION: Nebulized ASCs moderately improved pulmonary function and delayed the onset of ARDS.


Subject(s)
Acute Lung Injury , Mesenchymal Stem Cells , Respiratory Distress Syndrome , Smoke Inhalation Injury , Sheep , Animals , Female , Smoke Inhalation Injury/therapy , Smoke Inhalation Injury/complications , Pulmonary Gas Exchange , Lung , Acute Lung Injury/therapy , Acute Lung Injury/complications , Smoke/adverse effects , Respiratory Distress Syndrome/etiology , Disease Models, Animal
9.
Appl Immunohistochem Mol Morphol ; 31(5): 311-317, 2023.
Article in English | MEDLINE | ID: mdl-37010513

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a life-threatening condition due to acute lung injury (ALI), characterized by rapid-onset respiratory failure, leading to the clinical manifestations of poor lung compliance, severe hypoxemia, and dyspnea. ARDS/ALI has many causes, most commonly related to infections (sepsis, pneumonia), traumas, and multiple transfusions. The objective of this study is to assess the performance of postmortem anatomopathological examination in identifying etiological agents associated with ARDS or ALI in deceased patients from the State of São Paulo from 2017 to 2018. A retrospective cross-sectional study was performed based on the final outcome obtained by histopathology, histochemical, and immunohistochemical examination for ARDS/ALI differential diagnosis at the Pathology Center of the Adolfo Lutz Institute in São Paulo, Brazil. Of the 154 patients clinically diagnosed with ARDS or ALI, 57% tested positive for infectious agents, and the most frequent outcome was influenza A/H1N1 virus infection. In 43% of cases, no etiologic agent was identified. The opportunity to establish a diagnosis, identify particular infections, confirm a microbiological diagnosis, and uncover unanticipated etiologies is provided by postmortem pathologic analysis of ARDS. A molecular assessment could improve the diagnosis accuracy and lead to research into host responses and public health measures.


Subject(s)
Acute Lung Injury , Influenza A Virus, H1N1 Subtype , Respiratory Distress Syndrome , Humans , Retrospective Studies , Cross-Sectional Studies , Brazil , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology , Acute Lung Injury/complications
10.
ACS Biomater Sci Eng ; 9(5): 2716-2730, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37079432

ABSTRACT

If not properly managed, acute lung injuries, either through direct or indirect causes, have the potential to present serious risk for many patients worldwide. One of the mechanisms for the transition from acute lung injury (ALI) to the more serious acute respiratory distress syndrome (ARDS) is the deactivation of the native lung surfactant by injury-induced infiltrates to the alveolar space. Currently, there are no surfactant replacement therapies that are used to treat ALI and subsequent ARDS. In this paper, we present an indepth efficacy study of using a novel polymer lung surfactant (PLS, composed of poly(styrene-block-ethylene glycol) (PS-PEG) block copolymer micelles), which has unique properties compared to other tested surfactant replacements, in two different mouse models of lung injury. The results demonstrate that pharyngeal administration of PLS after the instillation of either acid (HCl) or lipopolysaccharide (LPS) can decrease the severity of lung injury as measured by multiple injury markers.


Subject(s)
Acute Lung Injury , Pulmonary Surfactants , Respiratory Distress Syndrome , Mice , Animals , Polymers/pharmacology , Polymers/therapeutic use , Lung , Pulmonary Surfactants/pharmacology , Pulmonary Surfactants/therapeutic use , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/etiology , Acute Lung Injury/drug therapy , Acute Lung Injury/complications
11.
Acta Neurol Belg ; 123(4): 1331-1337, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36922484

ABSTRACT

Acute lung injury or acute respiratory distress syndrome (ALI/ARDS) is a common complication after aneurysmal subarachnoid hemorrhage (aSAH), and is associated with worse neurologic outcomes and longer hospitalization. However, the effect of ALI/ARDS in SAH has not been well elucidated. The purpose of this study was to determine the incidence of ALI/ARDS in a cohort of patients with SAH and to determine the risk factors for ALI/ARDS and their impact on patient prognosis. We performed a retrospective analysis of 167 consecutive patients with aSAH enrolled. ALI/ARDS patients were rigorously adjudicated using North American-European Consensus Conference definition. Regression analyses were used to test the risk factors for ALI/ARDS in patients with SAH. A total of 167 patients fulfilled the inclusion criteria, and 27% patients (45 of 167) developed ALI. Among all 45 ALI patients, 33 (20%, 33 of 167) patients met criteria for ARDS. On multivariate analysis, elderly patients, lower glasgow coma scale (GCS), higher Hunt-Hess grade, higher simplified acute physiology score (SAPS) II score, pre-existing pneumonia, gastric aspiration, hypoxemia, and tachypnea were the strongest risk factor for ALI/ARDS. Patients with ALI/ARDS showed worse clinical outcomes measured at 30 days. Development of ALI/ARDS was associated with a statistically significant increasing the odds of tracheostomy and hospital complications, and increasing duration of mechanical ventilation, intensive care unit (ICU) length and hospitalization stay. Development of ALI/ARDS is a severe complication of SAH and is associated with a poor clinical outcome, and further studies should focus on both prevention and management strategies specific to SAH-associated ALI/ARDS.


Subject(s)
Acute Lung Injury , Respiratory Distress Syndrome , Subarachnoid Hemorrhage , Humans , Aged , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/epidemiology , Retrospective Studies , Respiratory Distress Syndrome/epidemiology , Respiratory Distress Syndrome/etiology , Prognosis , Acute Lung Injury/etiology , Acute Lung Injury/complications
12.
Surg Today ; 53(8): 973-983, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36592222

ABSTRACT

PURPOSE: Systematic inflammation has been reported to contribute to cancer progression through various mechanisms; however, the exact mechanism is still the subject of research. In this study, we evaluated the influence of systematic inflammation on lung metastasis, using a murine abdominal sepsis model, and assessed its relationship with pneumonia after curative esophagectomy in patients with esophageal cancer. METHODS: We used a murine abdominal sepsis model given highly metastatic osteosarcoma, to reveal the mechanism of systematic inflammation and its potential for lung metastasis. The therapeutic effect of aspirin (ASA) in preventing distant metastasis was also investigated. Subsequently, we analyzed, retrospectively, the relationship between pneumonia and lung metastasis after esophagectomy in patients who underwent esophagectomy at Keio University between January, 2007 and October, 2020. RESULTS: Abdominal sepsis provoked lung injury in the acute phase. ASA inhibited the recruitment of neutrophils triggered by the lung injury, and it also suppressed lung metastasis. Our retrospective study revealed that lung metastasis was more frequent in patients with postoperative pneumonia. CONCLUSIONS: Postoperative acute lung injury is associated with a higher risk of lung metastasis. ASA may be a potential preoperative treatment for inhibiting lung metastasis by preventing the recruitment of neutrophils.


Subject(s)
Acute Lung Injury , Lung Neoplasms , Pneumonia , Sepsis , Humans , Animals , Mice , Retrospective Studies , Aspirin/pharmacology , Neutrophil Infiltration , Pneumonia/etiology , Pneumonia/prevention & control , Inflammation/prevention & control , Acute Lung Injury/prevention & control , Acute Lung Injury/complications , Neutrophils/pathology , Lung Neoplasms/pathology , Lung/pathology
13.
Resuscitation ; 182: 109651, 2023 01.
Article in English | MEDLINE | ID: mdl-36442595

ABSTRACT

AIM: Describe the lung injury patterns among patients presenting with refractory ventricular tachycardia/ventricular fibrillation out-of-hospital cardiac arrest (VT/VF OHCA) supported with veno-arterial extracorporeal membrane oxygenation (VA-ECMO) facilitated resuscitation. METHODS: In this retrospective single-center cohort study including VT/VF OHCA patients supported with VA ECMO, we compared OHCA characteristics, post-arrest computed tomography (CT) scans, ventilator parameters, and other lung-related pathology between survivors, patients who developed brain death, and those with other causes of death. RESULTS: Among 138 patients, 48/138 (34.8%) survived, 31/138 (22.4%) developed brain death, and 59/138 (42.7%) died of other causes. Successful extubation was achieved in 39/138 (28%) with a median time to extubation of 8.0 days (6.0, 11.0) in those who survived. Tracheostomy was required in 15/48 (31.3%) survivors. Chest CT obtained on all patients showed lung injury in at least one lung area in 124/135 (91.8%) patients, predominantly in the dependent posterior areas. There was no association between the number of affected areas and survival. Lung compliance was low on admission [26 (19,33) ml/cmH20], improved throughout hospitalization (p = 0.03), and recovered faster in survivors compared to those who died (p < 0.001). VA-ECMO allowed the use of lung-protective ventilation while maintaining normalized PaO2 and PaCO2. Patients treated with V-A ECMO and either IABP or Impella had lower pulmonary compliance and more affected areas on their CT compared to those treated with V-A ECMO alone. CONCLUSIONS: Lung injury is common among patients with refractory VT/VF OHCA requiring V-A ECMO, but imaging severity is not associated with survival. Reductions in lung compliance accompany post-arrest lung injury while compliance recovery is associated with survival.


Subject(s)
Acute Lung Injury , Cardiopulmonary Resuscitation , Extracorporeal Membrane Oxygenation , Out-of-Hospital Cardiac Arrest , Humans , Ventricular Fibrillation/therapy , Extracorporeal Membrane Oxygenation/methods , Cohort Studies , Retrospective Studies , Brain Death , Cardiopulmonary Resuscitation/methods , Acute Lung Injury/complications
14.
Pediatr Res ; 93(4): 1050-1056, 2023 03.
Article in English | MEDLINE | ID: mdl-35906303

ABSTRACT

BACKGROUND: Sepsis related acute lung injury (ALI) is established in adults but has not been investigated in premature infants. Herein, we used pulmonary severity score (PSS) trajectories and C-reactive protein (CRP) to examine the relation between sepsis and ALI in premature infants. METHODS: This retrospective study identified 211 sepsis and 123 rule out (RO) events in 443 infants born <31 weeks and <1500 grams. The PSS was calculated prior to, at the time of, and up to 1 week after each event. Initial and peak CRP values were collected for each event. RESULTS: PSS significantly increased at 0 h from baseline (-72h) and remained increased at all subsequent time points (all p < 0.002) in sepsis events. Mean PSS in sepsis episodes were also higher compared to RO events at +24 h, +48 h, +72 h, and +168 h (all p < 0.004). A positive correlation was noted between peak CRP values in sepsis events and PSS at 0 h, +24 h, +48 h, and +72 h. CONCLUSIONS: The temporal PSS trends and correlation with CRP levels observed in sepsis but not in RO events supports the hypothesis that neonatal sepsis is associated with ALI and contributes to the accumulating evidence that neonatal ARDS occurs. IMPACT: To evaluate pulmonary severity scores and c-reactive protein values over time to establish an association between preterm neonatal sepsis and acute lung injury (ALI). Though sepsis is well established as the most common indirect cause of ALI leading to acute respiratory distress syndrome (ARDS) in adults and pediatrics, this phenomenon remains undefined in neonates. This study validates the proposal by the Neonatal ARDS Project that ARDS also occurs in neonates by demonstrating acute and sustained changes in markers of pulmonary injury temporally related to a diagnosis of neonatal sepsis in preterm infants.


Subject(s)
Acute Lung Injury , Neonatal Sepsis , Respiratory Distress Syndrome, Newborn , Respiratory Distress Syndrome , Sepsis , Adult , Humans , Infant, Newborn , Child , Neonatal Sepsis/complications , Neonatal Sepsis/diagnosis , Retrospective Studies , C-Reactive Protein/analysis , Infant, Premature , Sepsis/complications , Sepsis/diagnosis , Acute Lung Injury/complications , Acute Lung Injury/diagnosis , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome, Newborn/complications , Respiratory Distress Syndrome, Newborn/diagnosis
15.
Eur Respir J ; 61(2)2023 02.
Article in English | MEDLINE | ID: mdl-36229053

ABSTRACT

INTRODUCTION: Acute lung injury (ALI) is a major cause of morbidity and mortality after intestinal ischaemia/reperfusion (I/R). The gut microbiota and its metabolic byproducts act as important modulators of the gut-lung axis. This study aimed to define the role of succinate, a key microbiota metabolite, in intestinal I/R-induced ALI progression. METHODS: Gut and lung microbiota of mice subjected to intestinal I/R were analysed using 16S rRNA gene sequencing. Succinate level alterations were measured in germ-free mice or conventional mice treated with antibiotics. Succinate-induced alveolar macrophage polarisation and its effects on alveolar epithelial apoptosis were evaluated in succinate receptor 1 (Sucnr1)-deficient mice and in murine alveolar macrophages transfected with Sucnr1-short interfering RNA. Succinate levels were measured in patients undergoing cardiopulmonary bypass, including intestinal I/R. RESULTS: Succinate accumulated in lungs after intestinal I/R, and this was associated with an imbalance of succinate-producing and succinate-consuming bacteria in the gut, but not the lungs. Succinate accumulation was absent in germ-free mice and was reversed by gut microbiota depletion with antibiotics, indicating that the gut microbiota is a source of lung succinate. Moreover, succinate promoted alveolar macrophage polarisation, alveolar epithelial apoptosis and lung injury during intestinal I/R. Conversely, knockdown of Sucnr1 or blockage of SUCNR1 in vitro and in vivo reversed the effects of succinate by modulating the phosphoinositide 3-kinase-AKT/hypoxia-inducible factor-1α pathway. Plasma succinate levels significantly correlated with intestinal I/R-related lung injury after cardiopulmonary bypass. CONCLUSION: Gut microbiota-derived succinate exacerbates intestinal I/R-induced ALI through SUCNR1-dependent alveolar macrophage polarisation, identifying succinate as a novel target for gut-derived ALI in critically ill patients.


Subject(s)
Acute Lung Injury , Gastrointestinal Microbiome , Reperfusion Injury , Mice , Animals , Succinic Acid/metabolism , Phosphatidylinositol 3-Kinases , RNA, Ribosomal, 16S/genetics , Acute Lung Injury/complications , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Reperfusion , Ischemia/complications , Mice, Inbred C57BL
16.
Biomed Pharmacother ; 156: 113961, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36411667

ABSTRACT

This study investigated the impacts of enteral cholecalciferol and/or intravenous calcitriol administration on the balance of cluster of differentiation 4-positive T cell subsets, the renin-angiotensin system (RAS), and the severity of acute lung injury (ALI) in obese mice with sepsis. Mice were fed a high-fat diet and then cecal ligation and puncture (CLP) was performed. Obese mice were divided into four sepsis groups: without vitamin D (VD) (S), with oral cholecalciferol 1 d before CLP (G), with intravenous calcitriol 1 h after CLP (V), and with both cholecalciferol before and intravenous calcitriol after CLP (GV). Mice were euthanized after CLP. The V and GV groups showed higher blood T helper (Th)1/Th2 and lower Th17/T regulatory (Treg) ratios than did the S and G groups. In the lungs, The V group had the lowest nuclear factor-κB and interleukin-1ß gene expressions among all groups 24 h post-CLP. In parallel, gene expressions of angiotensin type 2 receptor (AT2R), angiotensin-converting enzyme 2 (ACE2), and Mas receptor (MasR) were highest in the V group compared to other groups. The protein levels of MasR in the GV group and the AT2R/AT1R ratio in the V group were higher than those in the G and/or S groups. All of the VD-treated groups had lower injury scores than the S group. These findings suggest that calcitriol administration had more-pronounced impacts on regulating the homeostasis of Th/Treg cells and is prone to RAS-associated anti-inflammatory pathway in the lungs. However, both forms of VD attenuated sepsis-induced ALI in obese animals.


Subject(s)
Acute Lung Injury , CD4-Positive T-Lymphocytes , Sepsis , Animals , Mice , Acute Lung Injury/complications , Calcitriol/pharmacology , Homeostasis , Mice, Obese , Receptor, Angiotensin, Type 2/metabolism , Renin-Angiotensin System , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Vitamin D/pharmacology , Vitamins , CD4-Positive T-Lymphocytes/immunology
17.
Mol Immunol ; 152: 232-239, 2022 12.
Article in English | MEDLINE | ID: mdl-36379131

ABSTRACT

Acute lung injury is a severe complication of sepsis with high mortality in ICU. Increasing evidences have showed that Ibrutinib, a Bruton's Tyrosine kinase inhibitor, plays a critical role in numerous inflammation-related diseases. However, its therapeutic effect and mechanism in sepsis induced acute lung injury remain unclear. In this study, cecal ligation puncture (CLP) was performed on male C57BL/6 J mice to establish a mouse model of sepsis. Ibrutinib (50 mg/kg/d) was administered by gavage 1 day before CLP, once a day, for 3 consecutive days. on the fourth day mice were given one dose of ibrutinib 2 h before CLP induction, and another dose was given 24 h later. Histopathological examination of lung tissues was performed at 72 h. The levels of myeloperoxidase (MPO), interleukin (IL)- 6, TNF-α, IL-1ß and IL-18 in bronchoalveolar lavage fluid (BALF) were determined by ELISA. Western blotting was used to detect the expression of pyroptosis related proteins. The results showed that Ibrutinib treatment significantly improved the prognosis of mice and mitigated the lung histopathological injury and inflammatory response. Moreover, Ibrutinib significantly inhibited the expression of pyroptosis related proteins (NLRP3, Caspase-1, Gasdermin D (GSDMD), IL-1ß and IL-18) in the lung tissues of sepsis mice. In conclusion, our results suggest that Ibrutinib exerted protective effects against lung injury of septic mice and inhibited the activation of pyroptosis in lung tissue, which may be a potential treatment for sepsis induced lung injury.


Subject(s)
Acute Lung Injury , Sepsis , Animals , Mice , Male , Caspase 1 , NLR Family, Pyrin Domain-Containing 3 Protein , Interleukin-18 , Mice, Inbred C57BL , Acute Lung Injury/drug therapy , Acute Lung Injury/complications , Disease Models, Animal , Sepsis/complications , Interleukin-6
18.
Crit Care ; 26(1): 274, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36100846

ABSTRACT

Acute neuropsychiatric impairments occur in over 70% of patients with acute lung injury. Mechanical ventilation is a well-known precipitant of acute lung injury and is strongly associated with the development of acute delirium and anxiety phenotypes. In prior studies, we demonstrated that IL-6 mediates neuropathological changes in the frontal cortex and hippocampus of animals with mechanical ventilation-induced brain injury; however, the effect of systemic IL-6 inhibition on structural and functional acute neuropsychiatric phenotypes is not known. We hypothesized that a murine model of mechanical ventilation-induced acute lung injury (VILI) would induce neural injury to the amygdala and hippocampus, brain regions that are implicated in diverse neuropsychiatric conditions, and corresponding delirium- and anxiety-like functional impairments. Furthermore, we hypothesized that these structural and functional changes would reverse with systemic IL-6 inhibition. VILI was induced using high tidal volume (35 cc/kg) mechanical ventilation. Cleaved caspase-3 (CC3) expression was quantified as a neural injury marker and found to be significantly increased in the VILI group compared to spontaneously breathing or anesthetized and mechanically ventilated mice with 10 cc/kg tidal volume. VILI mice treated with systemic IL-6 inhibition had significantly reduced amygdalar and hippocampal CC3 expression compared to saline-treated animals and demonstrated amelioration in acute neuropsychiatric behaviors in open field, elevated plus maze, and Y-maze tests. Overall, these data provide evidence of a pathogenic role of systemic IL-6 in mediating structural and functional acute neuropsychiatric symptoms in VILI and provide preclinical justification to assess IL-6 inhibition as a potential intervention to ameliorate acute neuropsychiatric phenotypes following VILI.


Subject(s)
Acute Lung Injury , Delirium , Ventilator-Induced Lung Injury , Acute Lung Injury/complications , Acute Lung Injury/drug therapy , Animals , Delirium/complications , Disease Models, Animal , Interleukin-6 , Mice , Phenotype , Ventilator-Induced Lung Injury/pathology
19.
Korean J Anesthesiol ; 75(6): 518-529, 2022 12.
Article in English | MEDLINE | ID: mdl-35912428

ABSTRACT

BACKGROUND: Acute lung injury (ALI) is the most serious complication of subarachnoid hemorrhage (SAH). We investigated role of autophagy and inflammatory signaling pathways in lung damage and therapeutic effects of dexmedetomidine (DEX). METHODS: Fifty male Wistar rats were randomly divided into five groups: sham, SAH, SAH+ DEX5, SAH+DEX25, and SAH+DEX50. SAH was induced using endovascular perforation technique. All rats received mechanical ventilation for 60 minutes. At 2 and 24 h of SAH induction, SAH+DEX groups were treated with 5, 25, and 50 µg/kg of DEX, respectively. Histological ALI score and pulmonary edema were assessed after 48 h. Lung expression of LC3B, ATG3, p62, TLR4, TLR9, and NFκB was assessed using western blotting and quantitative PCR. Blood levels of IL-6, IL-1ß, IFN-γ, and TNFα were also assessed. RESULTS: SAH induced ALI and pulmonary edema, which were attenuated in SAH+DEX5 (P < 0.001 for both) and SAH+DEX25 groups (P = 0.001 and P < 0.001 for ALI and edema, respectively). Lung expressions of LC3B and ATG3 were upregulated in SAH group, which was attenuated in SAH+DEX5 and SAH+DEX25 groups. Lung expressions of TLR4, TLR9, and NFκB were increased in SAH group, which was attenuated in SAH+DEX5 group. Blood IL-6 level was increased in SAH group and attenuated in SAH+DEX5 and SAH+DEX25 groups. Blood IFN-γ level was lower in SAH group than in sham group, and it was increased in SAH+DEX25 group. CONCLUSIONS: Low-dose DEX treatment after SAH may protect against ALI by disrupting pathological brain-lung crosstalk and alleviating autophagy flux and TLR-dependent inflammatory pathways.


Subject(s)
Acute Lung Injury , Dexmedetomidine , Pulmonary Edema , Subarachnoid Hemorrhage , Animals , Male , Rats , Acute Lung Injury/prevention & control , Acute Lung Injury/complications , Autophagy , Dexmedetomidine/pharmacology , Interleukin-6/therapeutic use , NF-kappa B/metabolism , Pulmonary Edema/prevention & control , Pulmonary Edema/complications , Rats, Wistar , Signal Transduction , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism
20.
Commun Biol ; 5(1): 543, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35668098

ABSTRACT

Sepsis-induced acute lung injury (ALI) is a serious sepsis complication and the prevailing cause of death. Circulating plasma exosomes might exert a key role in regulating intercellular communication between immunological and structural cells, as well as contributing to sepsis-related organ damage. However, the molecular mechanisms by which exosome-mediated intercellular signaling exacerbate ALI in septic infection remains undefined. Therefore, we investigated the effect of macrophage-derived exosomal APN/CD13 on the induction of epithelial cell necrosis. Exosomal APN/CD13 levels in the plasma of septic mice and patients with septic ALI were found to be higher. Furthermore, increased plasma exosomal APN/CD13 levels were associated with the severity of ALI and fatality in sepsis patients. We found remarkably high expression of APN/CD13 in exosomes secreted by LPS-stimulated macrophages. Moreover, c-Myc directly induced APN/CD13 expression and was packed into exosomes. Finally, exosomal APN/CD13 from macrophages regulated necroptosis of lung epithelial cells by binding to the cell surface receptor TLR4 to induce ROS generation, mitochondrial dysfunction and NF-κB activation. These results demonstrate that macrophage-secreted exosomal APN/CD13 can trigger epithelial cell necroptosis in an APN/CD13-dependent manner, which provides insight into the mechanism of epithelial cell functional disorder in sepsis-induced ALI.


Subject(s)
Acute Lung Injury , Sepsis , Acute Lung Injury/complications , Animals , CD13 Antigens/pharmacology , Epithelial Cells , Humans , Lung , Macrophages , Mice , Necroptosis , Sepsis/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...