1
|
Linden K, Schmandt M, Muders T, Theuerkauf N, Schewe JC, Herberg U, Putensen C, Ehrentraut SF, Kreyer S. Estimation of Cardiac Output Under Veno-Venous Extracorporeal Membrane Oxygenation: Comparing Thermodilution Methods to 3D Echocardiography. ASAIO J 2024:00002480-990000000-00533. [PMID: 39074443 DOI: 10.1097/mat.0000000000002283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
Thermodilution methods to determine cardiac output (CO) may be affected by veno-venous extracorporeal membrane oxygenation (ECMO). We compared CO estimations by pulmonary arterial thermodilution using a pulmonary arterial catheter (COPAC), transpulmonary thermodilution (COTPTD), and three-dimensional echocardiography (3DEcho) (CO3DEcho) in 18 patients under veno-venous ECMO. Comparisons between CO3DEcho and COPAC, and COTPTD were performed using correlation statistics and Bland-Altman analysis. Blood flow on ECMO support ranged from 4.3 to 5.8 L/min (median 4.9 L/min). Cardiac output measured with three-dimensional echocardiography was 5.2 L/min (3.8/5.9), COPAC was 7.3 L/min (5.9/7.9), and COTPTD was 7.3 L/min (6/8.2) (median [25%/75% percentile]). Bland-Altman analysis of CO3DEcho and COPAC revealed a mean bias of -2.06 L/min, with limits of agreement from -4.16 to 0.04 L/min. Bland-Altman analysis of CO3DEcho and COTPTD revealed a mean bias of -2.22 L/min, with limits of agreement from -4.18 to -0.25 L/min. We found a negative mean bias and negative limits of agreement between CO3DEcho and COPAC/COTPTD. We concluded an influence on the estimation of CO by thermodilution under ECMO most likely due to loss of indicator resulting in an overestimation of CO. Clinicians should consider this when monitoring thermodilution-based CO under ECMO.
Collapse
Affiliation(s)
- Katharina Linden
- From the Department of Pediatric Cardiology, University Hospital Bonn, Bonn, Germany
- Department of Pediatric Cardiology, University Hospital Aachen, Aachen, Germany
| | - Mathias Schmandt
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Thomas Muders
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Nils Theuerkauf
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Jens-Christian Schewe
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Medical Centre Rostock, Rostock, Germany
| | - Ulrike Herberg
- From the Department of Pediatric Cardiology, University Hospital Bonn, Bonn, Germany
- Department of Pediatric Cardiology, University Hospital Aachen, Aachen, Germany
| | - Christian Putensen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Stefan Felix Ehrentraut
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Stefan Kreyer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
2
|
Rehman TA, John K, Maslow A. Protective Lung Ventilation: What Do We Know?-"In An Investigation, Details Matter"-Jack Reacher TV Series. J Cardiothorac Vasc Anesth 2023; 37:2572-2576. [PMID: 37423839 PMCID: PMC10264327 DOI: 10.1053/j.jvca.2023.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 07/11/2023]
Affiliation(s)
- T A Rehman
- Department of Anesthesiology, Beth Israel Deaconess Medical Center, Boston, MA
| | - K John
- Department of Anesthesiology, Rhode Island Hospital, Providence, RI
| | - A Maslow
- Department of Anesthesiology, Rhode Island Hospital, Providence, RI.
| |
Collapse
|
3
|
Yeom R, Gorgone M, Malinovic M, Panzica P, Maslow A, Augoustides JG, Marchant BE, Fernando RJ, Nampi RG, Pospishil L, Neuburger PJ. Surgical Aortic Valve Replacement in a Patient with Very Severe Chronic Obstructive Pulmonary Disease. J Cardiothorac Vasc Anesth 2023; 37:2335-2349. [PMID: 37657996 DOI: 10.1053/j.jvca.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 09/03/2023]
Affiliation(s)
- Richard Yeom
- Department of Anesthesiology, Westchester Medical Center, Valhalla, NY
| | - Michelle Gorgone
- Department of Anesthesiology, Rhode Island Hospital, Providence, RI
| | - Matea Malinovic
- Department of Anesthesiology, Westchester Medical Center, Valhalla, NY
| | - Peter Panzica
- Department of Anesthesiology, Westchester Medical Center, Valhalla, NY
| | - Andrew Maslow
- Department of Anesthesiology, Rhode Island Hospital, Providence, RI
| | - John G Augoustides
- Department of Anesthesiology and Critical Care, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Bryan E Marchant
- Department of Anesthesiology, Cardiothoracic and Critical Care Sections, Wake Forest University School of Medicine, Winston Salem, NC
| | - Rohesh J Fernando
- Department of Anesthesiology, Cardiothoracic Section, Wake Forest University School of Medicine, Medical Center Boulevard, Winston Salem, NC.
| | - Robert G Nampi
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, NYU Grossman School of Medicine, New York, NY
| | - Liliya Pospishil
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, NYU Grossman School of Medicine, New York, NY
| | - Peter J Neuburger
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
4
|
Freundlich RE, Li G, Leis A, Engoren M. Factors Associated With Initiation of Mechanical Ventilation in Patients With Sepsis: Retrospective Observational Study. Am J Crit Care 2023; 32:358-367. [PMID: 37652887 PMCID: PMC10577809 DOI: 10.4037/ajcc2023299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
BACKGROUND Patients with sepsis are at risk for mechanical ventilation. This study aimed to identify risk factors for initiation of mechanical ventilation in patients with sepsis and assess whether these factors varied with time. METHODS Data from the electronic health record were used to model risk factors for initiation of mechanical ventilation after the onset of sepsis. A time-varying Cox model was used to study factors that varied with time. RESULTS Of 35 020 patients who met sepsis criteria, 28 747 were eligible for inclusion. Mechanical ventilation was initiated within 30 days after sepsis onset in 3891 patients (13.5%). Factors that were independently associated with increased likelihood of receipt of mechanical ventilation were race (White: adjusted hazard ratio [HR], 1.59; 95% CI, 1.39-1.83; other/unknown: adjusted HR, 1.97; 95% CI, 1.54-2.52), systemic inflammatory response syndrome (adjusted HR [per point], 1.23; 95% CI, 1.17-1.28), Sequential Organ Failure Assessment score (adjusted HR [per point], 1.28; 95% CI, 1.26-1.31), and congestive heart failure (adjusted HR, 1.30; 95% CI, 1.17-1.45). Hazard ratios decreased with time for Sequential Organ Failure Assessment score and congestive heart failure and varied with time for 4 comorbidities and 3 culture results. CONCLUSIONS The risk for mechanical ventilation associated with different factors varied with time after sepsis onset, increasing for some factors and decreasing for others. Through a better understanding of risk factors for initiation of mechanical ventilation in patients with sepsis, targeted interventions may be tailored to high-risk patients.
Collapse
Affiliation(s)
- Robert E Freundlich
- Robert E. Freundlich is an associate professor, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gen Li
- Gen Li is a senior statistical analyst, Department of Anesthesiology, Vanderbilt University Medical Center
| | - Aleda Leis
- Aleda Leis is a research investigator, Department of Epidemiology, University of Michigan, Ann Arbor
| | - Milo Engoren
- Milo Engoren is a professor, Department of Anesthesiology, University of Michigan
| |
Collapse
|
5
|
Dong D, Jing C, Zong Y, Wang Y, Ren J. Effect of different titration methods on right heart function and prognosis in patients with acute respiratory distress syndrome. Heart Lung 2023; 61:127-135. [PMID: 37263145 DOI: 10.1016/j.hrtlng.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/17/2023] [Accepted: 05/21/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a common disease in intensive critical care(ICU), and the use of positive end-expiratory pressure(PEEP) during mechanical ventilation can increase the right heart afterload and eventually cause right heart dysfunction. For these factors causing acute cor pulmonale(ACP), especially inappropriate mechanical ventilation settings, it is important to explore the effect of PEEP on right heart function. OBJECTIVE To investigate the effects of three titration methods on right heart function and prognosis in patients with ARDS. METHODS Observational, prospective study in which ARDS patients were enrolled into three distinct PEEP-titration strategies groups: guide, transpulmonary pressure-oriented and driving pressure-oriented. Prognostic indicators, right heart systolic and diastolic echocardiographic function indices, ventilatory parameters, blood gas analysis results, and respiratory mechanics Monitoring indices were collated and analyzed statistically by STATA 15 software. RESULTS A total of 62 ARDS patients were enrolled into guide (G) group (n=40) for whom titrated PEEP values were 9±2cm H2O, driving pressure-oriented (DPO) group (n=12) with titrated PEEP values of 10±2cm H2O and transpulmonary pressure-oriented (TPO) group (n=10) with titrated PEEP values of 12±3cm H2O. Values were significantly higher for TPO than for G (p=0.616) or DPO (p=0.011). Compliance was significantly increased after 72 h in the TPO and DPO groups compared with the G group (p<0.001). Mean airway pressure at end-inspiratory obstruction (p=0.047), tricuspid annular plane systolic excursion (TAPSE, p<0.001) and right ventricular area change fraction (RVFAC, p=0.049) were all higher in the TPO and DPO groups than in the G group. E/A indices were significantly better in the TPO group than in the G or DPO groups (p=0.046). No significant differences in 28 day mortality were found among the three groups. Multivariate logistic regression analysis revealed that lung compliance and transpulmonary pressure-oriented PEEP titration method was negatively correlated to the increase in right ventricular systolic dysfunction. CONCLUSION Transpulmonary pressure-oriented PEEP titration improves oxygenation and pulmonary function and causes less right heart strain when compared to other PEEP-titration methods during mechanical ventilation of ARDS patients.
Collapse
Affiliation(s)
- Daoran Dong
- Department of ICU, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Chengqiao Jing
- Department of ICU, Shaanxi Provincial People's Hospital, Xi'an, China.
| | - Yuan Zong
- Department of ICU, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yan Wang
- Department of ICU, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jiawei Ren
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
6
|
Pienkos SM, Moore AR, Guan J, Levitt JE, Matthay MA, Baron RM, Conlon J, McAuley DF, O'Kane CM, Rogers AJ. Effect of total cholesterol and statin therapy on mortality in ARDS patients: a secondary analysis of the SAILS and HARP-2 trials. Crit Care 2023; 27:126. [PMID: 36978134 PMCID: PMC10053133 DOI: 10.1186/s13054-023-04387-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/28/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Two acute respiratory distress syndrome (ARDS) trials showed no benefit for statin therapy, though secondary analyses suggest inflammatory subphenotypes may have a differential response to simvastatin. Statin medications decrease cholesterol levels, and low cholesterol has been associated with increased mortality in critical illness. We hypothesized that patients with ARDS and sepsis with low cholesterol could be harmed by statins. METHODS Secondary analysis of patients with ARDS and sepsis from two multicenter trials. We measured total cholesterol from frozen plasma samples obtained at enrollment in Statins for Acutely Injured Lungs from Sepsis (SAILS) and Simvastatin in the Acute Respiratory Distress Syndrome (HARP-2) trials, which randomized subjects with ARDS to rosuvastatin versus placebo and simvastatin versus placebo, respectively, for up to 28 days. We compared the lowest cholesterol quartile (< 69 mg/dL in SAILS, < 44 mg/dL in HARP-2) versus all other quartiles for association with 60-day mortality and medication effect. Fisher's exact test, logistic regression, and Cox Proportional Hazards were used to assess mortality. RESULTS There were 678 subjects with cholesterol measured in SAILS and 509 subjects in HARP-2, of whom 384 had sepsis. Median cholesterol at enrollment was 97 mg/dL in both SAILS and HARP-2. Low cholesterol was associated with higher APACHE III and shock prevalence in SAILS, and higher Sequential Organ Failure Assessment score and vasopressor use in HARP-2. Importantly, the effect of statins differed in these trials. In SAILS, patients with low cholesterol who received rosuvastatin were more likely to die (odds ratio (OR) 2.23, 95% confidence interval (95% CI) 1.06-4.77, p = 0.02; interaction p = 0.02). In contrast, in HARP-2, low cholesterol patients had lower mortality if randomized to simvastatin, though this did not reach statistical significance in the smaller cohort (OR 0.44, 95% CI 0.17-1.07, p = 0.06; interaction p = 0.22). CONCLUSIONS Cholesterol levels are low in two cohorts with sepsis-related ARDS, and those in the lowest cholesterol quartile are sicker. Despite the very low levels of cholesterol, simvastatin therapy seems safe and may reduce mortality in this group, though rosuvastatin was associated with harm.
Collapse
Affiliation(s)
- Shaun M Pienkos
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, 300 Pasteur Dr H3143, Stanford, CA, 94305, USA
| | - Andrew R Moore
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, 300 Pasteur Dr H3143, Stanford, CA, 94305, USA
| | - Jiazhen Guan
- Division of Pulmonary & Critical Care, Brigham and Women's Hosp, 15 Francis Street, Boston, MA, 02115, USA
| | - Joseph E Levitt
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, 300 Pasteur Dr H3143, Stanford, CA, 94305, USA
| | - Michael A Matthay
- Department of Medicine, Cardiovascular Research Institute, University of California, 35 Medical Center Way, San Francisco, CA, 94143, USA
| | - Rebecca M Baron
- Division of Pulmonary & Critical Care, Brigham and Women's Hosp, 15 Francis Street, Boston, MA, 02115, USA
| | - John Conlon
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Daniel F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, 97 Lisburn Road, Belfast, BT9 7BL, UK
- Regional Intensive Care Unit, Royal Victoria Hospital, 274 Grosvenor Rd, Belfast, BT12 6BA, UK
| | - Cecilia M O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Angela J Rogers
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, 300 Pasteur Dr H3143, Stanford, CA, 94305, USA.
| |
Collapse
|
7
|
Yang J, Chen X. SIRT6 attenuates LPS-induced inflammation and apoptosis of lung epithelial cells in acute lung injury through ACE2/STAT3/PIM1 signaling. Immun Inflamm Dis 2023; 11:e809. [PMID: 36988243 PMCID: PMC10022422 DOI: 10.1002/iid3.809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/10/2023] [Accepted: 02/18/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a severe and fatal respiratory disease. SIRT6 exerts pivotal activities in the process of lung diseases, but whether SIRT6 impacts ALI has not been covered. METHODS Lentivirus recombinant expressing vector SIRT6 gene (Lent-SIRT6) was constructed in mice, and there were control, lipopolysaccharide (LPS), LPS + Vehicle, and LPS + Lent SIRT6 groups. RT-qPCR and western blot detected SIRT6 expression in lung tissues. HE staining observed pathological alternations in lung tissues. Wet-to-dry ratio of the lungs was then measured. The cell count of bronchoalveolar lavage fluid (BALF) was evaluated. Serum inflammation was examined with enzyme-linked immunosorbent assay, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and western blot were to measure apoptosis. Western blot tested the expression of ACE2/STAT3/PIM1 signaling-associated factors. At the cellular level, LPS was used to induce lung epithelial cells BEAS-2B to establish cell injury models. SIRT6 was overexpressed and ACE2 expression was inhibited by cell transfection, and the mechanism of SIRT6 in LPS-induced lung injury model was further explored by Cell Counting Kit-8 (CCK-8), western blot, quantitative reverse-transcription polymerase chain reaction, TUNEL, and other techniques. RESULTS The results of animal experiments showed that SIRT6 overexpression could reduce LPS-induced lung pathological injury, pulmonary edema, and BALF cell ratio and attenuate LPS-induced inflammatory response and cell apoptosis. In the above process, ACE2, STAT3, p-STAT3, and PIM1 expression were affected. In cell experiments, SIRT6 expression was reduced in LPS-induced BEAS-2B cells. Inhibition of ACE2 expression could reverse the inhibitory effect of SIRT6 overexpression on ACE2/STAT3/PIM1 pathway, and cellular inflammatory response and apoptosis. CONCLUSION SIRT6 eased LPS-evoked inflammation and apoptosis of lung epithelial cells in ALI through ACE2/STAT3/PIM1 signaling.
Collapse
Affiliation(s)
- Juan Yang
- Department of Pediatric, Shandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Xing Chen
- Department of Pediatric, Shandong Provincial HospitalShandong UniversityJinanShandongChina
| |
Collapse
|
8
|
Zhang JB, Li LH, Zhu JQ, Zhou SF, Ma JH, Li ZQ, Jin XH, Lin XQ. Application of improved Glasgow coma scale score as switching point for sequential invasive-noninvasive mechanical ventilation on chronic obstructive pulmonary disease (COPD) with respiratory failure. Medicine (Baltimore) 2022; 101:e31857. [PMID: 36401492 PMCID: PMC9678540 DOI: 10.1097/md.0000000000031857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND To compare the efficacy and feasibility of using a modified Glasgow coma scale (GCS) score of 13 or 15 as the criterion for switching chronic obstructive pulmonary disease (COPD) patients with respiratory failure to sequential invasive-noninvasive ventilation. METHODS COPD patients with respiratory failure who had undergone endotracheal intubation and invasive mechanical ventilation (IMV) between June 2017 and June 2020 at 4 different hospitals in China were included. A total of 296 patients were randomly divided into 2 groups. In group A, the patients were extubated and immediately placed on noninvasive ventilation (NIV) when the modified GCS score reached 13. In group B, the same was done when the modified GCS score reached 15. RESULTS No significant differences in the mean blood pressure, oxygenation index, arterial partial pressure of oxygen, and arterial partial pressure of carbon dioxide were seen between groups A and B before extubation and 3 hours after NIV. The re-intubation times were also similar in the 2 groups. Compared to group B, the length of hospital stay, incidence of ventilator associated pneumonia, and time of invasive ventilation were all significantly lower in group A (P = .041, .001, <.001). CONCLUSION Using a modified GCS score of 13 as the criterion for switching from IMV to NIV can significantly reduce the duration of IMV, length of hospital stay, and incidence of ventilator associated pneumonia in COPD patients with respiratory failure.
Collapse
Affiliation(s)
- Jin-Bo Zhang
- Emergency Intensive Care Unit, Wenling Hospital Affiliated to Wenzhou Medical University, The First People’s Hospital of Wenling, Wenling, Zhejiang, China
| | - Li-Hong Li
- Infection Division, Wenling Hospital Affiliated to Wenzhou Medical University, The First People’s Hospital of Wenling, Wenling, Zhejiang, China
| | - Jin-Qiang Zhu
- Emergency Intensive Care Unit, Wenling Hospital Affiliated to Wenzhou Medical University, The First People’s Hospital of Wenling, Wenling, Zhejiang, China
| | - Shi-Fang Zhou
- Department of Emergency Care, Changsha Central Hospital, Changsha, Hunan, China
| | - Ji-Hong Ma
- Intensive Care Unit, First Affiliated Hospital of Wenzhou Medical University, Wenling, Zhejiang, China
| | - Zhi-Qiang Li
- Intensive Care Unit, The First People’s Hospital of Jingmen, Jingmen, Hubei, China
| | - Xiao-Hong Jin
- Emergency Intensive Care Unit, Wenling Hospital Affiliated to Wenzhou Medical University, The First People’s Hospital of Wenling, Wenling, Zhejiang, China
| | - Xiao-Qin Lin
- Department of Hepatopancreatobiliary Surgery, Wenling Hospital Affiliated to Wenzhou Medical University, The First People’s Hospital of Wenling, Wenling, Zhejiang, China
- * Correspondence: Xiao-Qin Lin, Department of Hepatopancreatobiliary Surgery, Wenling Hospital Affiliated to Wenzhou Medical University, The First People’s Hospital of Wenling, No. 333 Chuan an south Road, Wenling, Zhejiang 317500, China (e-mail: )
| |
Collapse
|
9
|
Zhou C, Chase JG. Low-cost structured light imaging of regional volume changes for use in assessing mechanical ventilation. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 226:107176. [PMID: 36228494 DOI: 10.1016/j.cmpb.2022.107176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/21/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Optimal setting of mechanical ventilators is critical for improving outcomes. Accurate, predictive lung mechanics models are effective in optimizing MV settings, but only at a global level as they cannot estimate regional lung volume ventilation to assess the potential of local distension or under-ventilation. This study presents a low-cost structured light system for non-contact high resolution chest motion measurement to estimate regional lung volume changes. METHODS The system consists of a structured light projector and two cameras. A new pattern is designed to extract motion from sub-regions of the chest surface, and an efficient feature is proposed to provide a fast and accurate correspondence matching between two views. Reconstruction of 3D surface points is based on the matched points and stereo method. Asymmetric distribution of tidal volume into left and right lungs is estimated based on reconstructed regional chest expansion. A proof-of-concept experiment using a dummy model and two test lungs connected to a ventilator to provide differential chest expansion is conducted under tidal volumes of 400 ml, 500 ml and 600 ml, with results compared to the widely-used SURF and ORB methods. RESULTS Compared to the SURF and ORB methods, the proposed method is more computationally efficient with ∼40% less computational time cost, and higher accuracy for dense point correspondence. Finally, the proposed method estimated the region lung volumes with the maximum error of 8 ml under 600 ml tidal volume, indicating a good accuracy. CONCLUSIONS Surface reconstruction results in a proof-of-concept experiment with differential chest expansion show good performance for the proposed pattern and method in extracting the key information for regional chest expansion. The proposed method is generalizable, with potential for use in other applications.
Collapse
Affiliation(s)
- Cong Zhou
- School of Civil Aviation, Northwestern Polytechnical University, China; Department of Mechanical Engineering, Centre for Bio-Engineering, University of Canterbury, Christchurch, New Zealand
| | - J Geoffrey Chase
- Department of Mechanical Engineering, Centre for Bio-Engineering, University of Canterbury, Christchurch, New Zealand.
| |
Collapse
|
10
|
Hao X, Wei H. LncRNA H19 alleviates sepsis-induced acute lung injury by regulating the miR-107/TGFBR3 axis. BMC Pulm Med 2022; 22:371. [PMID: 36180862 PMCID: PMC9524034 DOI: 10.1186/s12890-022-02091-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/27/2022] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE Acute lung injury (ALI) increases sepsis morbidity and mortality. LncRNA H19 plays a critical role in sepsis. miR-107 is highly-expressed and TGFβ type III receptor (TGFBR3) is poorly-expressed in sepsis, yet their roles in sepsis development require further investigation. This study aimed to investigate the mechanism of H19 in alleviating sepsis-induced ALI through the miR-107/TGFBR3 axis. METHODS Mice were intravenously injected with Ad-H19 adenovirus vector or control vector one week before establishing the mouse model of cecal ligation and puncture (CLP). Pulmonary microvascular endothelial cells (PMVECs) were transfected with oe-H19 or oe-NC plasmids and then stimulated by lipopolysaccharide (LPS). Lung injury was assessed via hematoxylin-eosin staining, measurement of wet-to-dry (W/D) ratio, and TUNEL staining. Levels of H19, miR-107, and TGFBR3 were determined by RT-qPCR. Apoptosis of PMVECs was evaluated by flow cytometry. Levels of Bax and Bcl-2 in lung tissues and PMVECs were measured using Western blot. Total protein concentration and the number of total cells, neutrophils, and macrophages in bronchoalveolar lavage fluid (BALF) were quantified. Levels of TNF-α, IL-1β, IL-6, and IL-10 in BALF, lung tissues, and PMVECs were measured by ELISA. Cross-linking relationships among H19, miR-107 and TGFBR3 were verified by dual-luciferase and RIP assays. RESULTS H19 was poorly-expressed in CLP-operated mice. H19 overexpression attenuated sepsis-induced ALI, which was manifested with complete alveolar structure, decreased lung injury score and lung W/D ratio, and inhibited apoptosis in CLP-operated mice, which was manifested with decreased number of TUNEL-positive cells and Bax level and increased Bcl-2 level. CLP-operated mice had increased concentration of total protein and number of total cells, neutrophils, and macrophages in BALF, which was nullified by H19 overexpression. H19 overexpression declined levels of TNF-α, IL-1β, and IL-6 and elevated IL-10 levels. H19 inhibited LPS-induced PMVEC apoptosis and pro-inflammatory cytokine production. H19 targeted TGFBR3 as the ceRNA of miR-107. miR-107 overexpression or silencing TGFBR3 partially averted the inhibition of H19 overexpression on LPS-induced PMVEC apoptosis and pro-inflammatory cytokine production. CONCLUSION LncRNA H19 inhibited LPS-induced PMVEC apoptosis and pro-inflammatory cytokine production and attenuated sepsis-induced ALI by targeting TGFBR3 as the ceRNA of miR-107.
Collapse
Affiliation(s)
- Xiuling Hao
- Department of Respiratory Medicine, East Hospital, The Second Hospital of Hebei Medical University, No. 80, Huanghe Avenue, East Development Zone, Shijiazhuang City, 050000, Hebei Province, People's Republic of China
| | - Huiqiang Wei
- Department of Respiratory Medicine, East Hospital, The Second Hospital of Hebei Medical University, No. 80, Huanghe Avenue, East Development Zone, Shijiazhuang City, 050000, Hebei Province, People's Republic of China.
| |
Collapse
|
11
|
Andrews P, Shiber J, Madden M, Nieman GF, Camporota L, Habashi NM. Myths and Misconceptions of Airway Pressure Release Ventilation: Getting Past the Noise and on to the Signal. Front Physiol 2022; 13:928562. [PMID: 35957991 PMCID: PMC9358044 DOI: 10.3389/fphys.2022.928562] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/21/2022] [Indexed: 12/16/2022] Open
Abstract
In the pursuit of science, competitive ideas and debate are necessary means to attain knowledge and expose our ignorance. To quote Murray Gell-Mann (1969 Nobel Prize laureate in Physics): "Scientific orthodoxy kills truth". In mechanical ventilation, the goal is to provide the best approach to support patients with respiratory failure until the underlying disease resolves, while minimizing iatrogenic damage. This compromise characterizes the philosophy behind the concept of "lung protective" ventilation. Unfortunately, inadequacies of the current conceptual model-that focuses exclusively on a nominal value of low tidal volume and promotes shrinking of the "baby lung" - is reflected in the high mortality rate of patients with moderate and severe acute respiratory distress syndrome. These data call for exploration and investigation of competitive models evaluated thoroughly through a scientific process. Airway Pressure Release Ventilation (APRV) is one of the most studied yet controversial modes of mechanical ventilation that shows promise in experimental and clinical data. Over the last 3 decades APRV has evolved from a rescue strategy to a preemptive lung injury prevention approach with potential to stabilize the lung and restore alveolar homogeneity. However, several obstacles have so far impeded the evaluation of APRV's clinical efficacy in large, randomized trials. For instance, there is no universally accepted standardized method of setting APRV and thus, it is not established whether its effects on clinical outcomes are due to the ventilator mode per se or the method applied. In addition, one distinctive issue that hinders proper scientific evaluation of APRV is the ubiquitous presence of myths and misconceptions repeatedly presented in the literature. In this review we discuss some of these misleading notions and present data to advance scientific discourse around the uses and misuses of APRV in the current literature.
Collapse
Affiliation(s)
- Penny Andrews
- R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joseph Shiber
- University of Florida College of Medicine, Jacksonville, FL, United States
| | - Maria Madden
- R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Gary F. Nieman
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Luigi Camporota
- Department of Adult Critical Care, Guy’s and St Thomas’ NHS Foundation Trust, Health Centre for Human and Applied Physiological Sciences, London, United Kingdom
| | - Nader M. Habashi
- R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Lou Y, Huang Z, Wu H, Zhou Y. Tranilast attenuates lipopolysaccharide‑induced lung injury via the CXCR4/JAK2/STAT3 signaling pathway. Mol Med Rep 2022; 26:220. [PMID: 35583012 PMCID: PMC9175269 DOI: 10.3892/mmr.2022.12736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/15/2021] [Indexed: 12/02/2022] Open
Abstract
It has been reported that the expression of C-X-C motif chemokine receptor 4 (CXCR4) is increased in patients with lung injury, while CXCR4 downregulation can improve sepsis-induced lung injury. Previous studies have shown that tranilast can inhibit CXCR4 mRNA expression. Therefore, the present study aimed to investigate whether tranilast could protect against lipopolysaccharide (LPS)-induced lung injury via the CXCR4/Janus kinase 2 (JAK2)/STAT3 signaling pathway. A Cell Counting Kit-8 assay was performed to evaluate the effect of different concentrations of tranilast on the viability of LPS-induced BEAS-2B cells. The mRNA and protein expression levels of the inflammatory factors, TNFα, IL-1β, IL-6, cytochrome c oxidase subunit II and inducible nitric oxide synthase were detected using reverse transcription-quantitative PCR and western blot analysis, respectively. In addition, the cell apoptosis rate and the expression levels of apoptosis-related proteins were analyzed using a TUNEL staining assay and western blot analysis, respectively. The expression levels of the CXCR4/JAK2/STAT3 signaling pathway-related proteins were also determined using western blot analysis. Furthermore, the effects of tranilast on cell viability, inflammation and apoptosis were also evaluated in LPS-stimulated BEAS-2B cells following CXCR4 overexpression, which were pre-treated with tranilast. The results demonstrated that tranilast could alleviate LPS-induced cell viability, the secretion of inflammatory cytokines and cell apoptosis. In addition, cell treatment with tranilast inhibited the expression of CXCR4/JAK2/STAT3 signaling-related proteins in LPS-induced BEAS-2B cells. Following CXCR4 overexpression, the alleviating effect of tranilast on cell viability, inflammatory response and apoptosis was notably attenuated. Overall, the current study suggested that tranilast could attenuate LPS-induced lung injury via the CXCR4/JAK2/STAT3 signaling pathway, suggesting that tranilast could be considered as a promising agent for treating sepsis-induced acute lung injury.
Collapse
Affiliation(s)
- Yufeng Lou
- Department of Emergency, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Zhenrong Huang
- Department of Emergency, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Hui Wu
- Department of Emergency, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Yun Zhou
- Department of Emergency, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| |
Collapse
|
13
|
Wang R, Dai H. Association of platelet count with all-cause mortality from acute respiratory distress syndrome: A cohort study. J Clin Lab Anal 2022; 36:e24378. [PMID: 35358347 PMCID: PMC9102613 DOI: 10.1002/jcla.24378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background The purpose of this study was to investigate whether platelet count was associated with mortality in acute respiratory distress syndrome (ARDS) patients. Methods We analyzed patients with ARDS from Multi‐parameter Intelligent Monitoring in Intensive Care Database III (MIMIC‐III). Platelet count was measured at the time of intensive care unit (ICU) admission. The cox proportional hazard model and subgroup analysis were used to determine the relationship between the platelet count and mortality of ARDS, as well as the consistency of its association. The primary outcome of this study was 365‐day mortality from the date of ICU admission. Result This study enrolled a total of 395 critically ill patients with ARDS. After adjustment for age, gender and ethnicity, the multivariate cox regression model showed that the hazard ratios (HRs) (95% confidence intervals [CIs]) of platelet count <192 × 109/L and >296 × 109/L were 2.08 (1.43, 3.04) and 1.35 (0.91, 2.01), respectively, compared with the reference (192–296 ×109/L). After adjusting for confounding factors, lower platelet count (<192 × 109/L) was associated with increased mortality (adjusted HR, 1.71; 95% CI 1.06–2.76, p = 0.0284). However, there was no similar trend in the 30‐day (adjusted HR,1.02; 95% CI 0.54–1.94) or 90‐day (adjusted HR, 1.65; 95% CI 0.94–2.89) mortality. In the subgroup analysis, lower platelet count showed significant interactions with specific populations (p interaction = 0.0413), especially in patients with atrial fibrillation. Conclusion Taken together, our analysis showed that platelet count is an independent predictor of mortality in critically ill patients with ARDS.
Collapse
Affiliation(s)
- Rennv Wang
- Emergency Department, Affiliated Zhejiang Hospital of Zhejiang University School of Medical, Hangzhou, Zhejiang, China
| | - Haiwen Dai
- Emergency Department, Affiliated Zhejiang Hospital of Zhejiang University School of Medical, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Liu J, Fan G, Tao N, Sun T. Role of Pyroptosis in Respiratory Diseases and its Therapeutic Potential. J Inflamm Res 2022; 15:2033-2050. [PMID: 35370413 PMCID: PMC8974246 DOI: 10.2147/jir.s352563] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
Pyroptosis is an inflammatory type of regulated cell death that is dependent on inflammasome activation and downstream proteases such as caspase-1 or caspase 4/5/11. The main executors are gasdermins, which have an inherent pore-forming function on the membrane and release inflammatory cytokines, such as interleukin (IL)-1β, IL-18 and high mobility group box 1. Emerging evidence demonstrates that pyroptosis is involved in the pathogenesis of various pulmonary diseases. In this review, we mainly discuss the biological mechanisms of pyroptosis, explore the relationship between pyroptosis and respiratory diseases, and discuss emerging therapeutic strategies for respiratory diseases.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Respiratory Medicine and Critical Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Beijing, People’s Republic of China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Guoqing Fan
- Department of Respiratory Medicine and Critical Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Beijing, People’s Republic of China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Ningning Tao
- Department of Respiratory Medicine and Critical Care, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Tieying Sun
- Department of Respiratory Medicine and Critical Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Beijing, People’s Republic of China
- Correspondence: Tieying Sun, Department of Respiratory Medicine and Critical Care, Beijing Hospital, Dongcheng District, Beijing, 100730, People’s Republic of China, Tel +86 15153169108, Email
| |
Collapse
|
15
|
Glibenclamide Alleviates LPS-Induced Acute Lung Injury through NLRP3 Inflammasome Signaling Pathway. Mediators Inflamm 2022; 2022:8457010. [PMID: 35185385 PMCID: PMC8856806 DOI: 10.1155/2022/8457010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/19/2022] [Indexed: 11/17/2022] Open
Abstract
Glibenclamide displays an anti-inflammatory response in various pulmonary diseases, but its exact role in lipopolysaccharide- (LPS-) induced acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) remains unknown. Herein, we aimed to explore the effect of glibenclamide in vivo and in vitro on the development of LPS-induced ALI in a mouse model. LPS stimulation resulted in increases in lung injury score, wet/dry ratio, and capillary permeability in lungs, as well as in total protein concentration, inflammatory cells, and inflammatory cytokines including IL-1β, IL-18 in bronchoalveolar lavage fluid (BALF), and lung tissues, whereas glibenclamide treatment reduced these changes. Meanwhile, the increased proteins of NLRP3 and Caspase-1/p20 after LPS instillation in lungs were downregulated by glibenclamide. Similarly, in vitro experiments also found that glibenclamide administration inhibited the LPS-induced upregulations in cytokine secretions of IL-1β and IL-18, as well as in the expression of components in NLRP3 inflammasome in mouse peritoneal macrophages. Of note, glibenclamide had no effect on the secretion of TNF-α in vivo nor in vitro, implicating that its anti-inflammatory effect is relatively specific to NLRP3 inflammasome. In conclusion, glibenclamide alleviates the development of LPS-induced ALI in a mouse model via inhibiting the NLRP3/Caspase-1/IL-1β signaling pathway, which might provide a new strategy for the treatment of LPS-induced ALI.
Collapse
|
16
|
Ge C, Liu J, Fu Y, Jia L, Long L, Dong S. MicroRNA-21 protects against sepsis-induced acute lung injury by targeting phosphatase and tensin homolog in mice. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221120978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction: Sepsis can cause acute lung injury (ALI), one of the leading causes of death in critically ill patients. The underlying mechanisms of sepsis-induced acute lung injury include excessive inflammation, oxidative stress, cell apoptosis, pulmonary edema, and lung tissue dysfunction. Recent studies have shown that miRNA-21 (miR-21) plays a vital role in sepsis-induced acute kidney injury. Relatively few studies have focused on the protective effects of ALI. This study aimed to determine the potential role of miR-21 in sepsis-induced ALI. Methods: We performed quantitative real-time polymerase chain reaction in a septic mouse model induced by cecal ligation and puncture (CLP) and found that miR-21 expression was upregulated. We then transfected the miR-21 precursor to upregulate miR-21 expression and miR-21 inhibitor to downregulate miR-21 expression. The sham group was exposed only to the cecum. ALI was induced by CLP, and the pre-miR-21+ALI and anti-miR-21+ALI groups were treated with miR-21 precursor or miR-21 inhibitor in the caudal vein before CLP. Pre-miR-21+ALI+PTEN inhibition (Pre-miR-21+ALI+PI) and anti-miR-21+ALI+PTEN inhibition (Anti-miR-21+ALI+PI) groups were treated with PTEN inhibition into the caudal vein after miR-21 transfection. Inflammatory cytokines, oxidative stress indicators, lung tissue cell apoptosis, oxygenation index (OI), lung wet/dry weight ratio, and lung pathological changes in the lung were observed in each group. Results: Compared with ALI mice, inflammatory response, oxidative stress indicators, lung tissue cell apoptosis, and the degree of lung injury were remarkably alleviated in Pre-miR-21+ALI mice and aggravated in Anti-miR-21+ALI mice. Western blot analysis showed that phosphatase and tensin homolog (PTEN) protein expression was decreased in CLP-treated mics. PTEN protein expression was decreased in the Pre-miR-21+ALI group but increased in the Anti-miR-21+ALI group. Moreover, the effect of miR-21 on anti-inflammatory, anti-oxidative stress, and anti-apoptosis enhanced after PTEN inhibition. Conclusion: This study revealed that miR-21 has a protective effect in sepsis-induced ALI by regulating PTEN in mice.
Collapse
Affiliation(s)
- Chen Ge
- Department of Intensive Medicine, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Junhang Liu
- Department of Orthopaedics Surgery, Children’s Hospital of Hebei, Shijiazhuang, P.R. China
| | - You Fu
- Department of Intensive Medicine, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Lijing Jia
- Department of Intensive Medicine, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Ling Long
- Department of Intensive Medicine, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Shimin Dong
- Department of Emergency, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, P.R. China
| |
Collapse
|
17
|
Kolomaznik M, Mikolka P, Hanusrichterova J, Kosutova P, Matasova K, Mokra D, Calkovska A. N-Acetylcysteine in Mechanically Ventilated Rats with Lipopolysaccharide-Induced Acute Respiratory Distress Syndrome: The Effect of Intravenous Dose on Oxidative Damage and Inflammation. Biomedicines 2021; 9:biomedicines9121885. [PMID: 34944701 PMCID: PMC8698392 DOI: 10.3390/biomedicines9121885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 12/30/2022] Open
Abstract
Treatment of acute respiratory distress syndrome (ARDS) is challenging due to its multifactorial aetiology. The benefit of antioxidant therapy was not consistently demonstrated by previous studies. We evaluated the effect of two different doses of intravenous (i.v.) N-acetylcysteine (NAC) on oxidative stress, inflammation and lung functions in the animal model of severe LPS-induced lung injury requiring mechanical ventilation. Adult Wistar rats with LPS (500 μg/kg; 2.2 mL/kg) were treated with i.v. NAC 10 mg/kg (NAC10) or 20 mg/kg (NAC20). Controls received saline. Lung functions, lung oedema, total white blood cell (WBC) count and neutrophils count in blood and bronchoalveolar lavage fluid, and tissue damage in homogenized lung were evaluated. NAC significantly improved ventilatory parameters and oxygenation, reduced lung oedema, WBC migration and alleviated oxidative stress and inflammation. NAC20 in comparison to NAC10 was more effective in reduction of oxidative damage of lipids and proteins, and inflammation almost to the baseline. In conclusion, LPS-instilled and mechanically ventilated rats may be a suitable model of ARDS to test the treatment effects at organ, systemic, cellular and molecular levels. The results together with literary data support the potential of NAC in ARDS.
Collapse
Affiliation(s)
- Maros Kolomaznik
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.K.); (P.K.)
| | - Pavol Mikolka
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (P.M.); (J.H.); (D.M.)
| | - Juliana Hanusrichterova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (P.M.); (J.H.); (D.M.)
| | - Petra Kosutova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.K.); (P.K.)
| | - Katarina Matasova
- Clinic of Neonatology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and Martin University Hospital, 03601 Martin, Slovakia;
| | - Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (P.M.); (J.H.); (D.M.)
| | - Andrea Calkovska
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (P.M.); (J.H.); (D.M.)
- Correspondence: ; Tel.: +421-43-2633-411
| |
Collapse
|
18
|
Liu W, Li Y, Bo L, Li C, Jin F. Positive regulation of TFEB and mitophagy by PGC-1α to alleviate LPS-induced acute lung injury in rats. Biochem Biophys Res Commun 2021; 577:1-5. [PMID: 34482051 DOI: 10.1016/j.bbrc.2021.08.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 11/27/2022]
Abstract
AIM OF THE STUDY Acute lung injury (ALI) exhibits the features of noncardiogenic pulmonary edema and acute inflammatory process, and it also displays significant morbidity and mortality rates. This work focused on identifying how overexpression of PPARγ coactivator 1α (PGC-1α) positively regulated TFEB and mitophagy for resisting the lipopolysaccharide (LPS)-mediated ALI. MATERIALS AND METHODS The levels of autophagic proteins and inflammatory factors in LPS-induced ALI rats and primary type II alveolar epithelial cells were measured, respectively. Lung wet/dry ratios were calculated. Protein co-immunoprecipitation of PGC-1α and TFEB was detected. To explore the interaction between TFEB and PGC-1α, a luciferase reporter assay was conducted. RESULTS The results showed that overexpression of PGC-1α decreases IL-1 and IL-6 but increases IL-10 in LPS-mediated ALI rats and type II alveolar epithelial cells (P < 0.05). Overexpression of PGC-1α can reduce lung edema in LPS-mediated ALI rats (P < 0.05). Overexpression of PGC-1α upregulates mitophagy-related proteins, such as TFEB, LC3B, Beclin, and LAMP1, and improves mitophagy in LPS-induced ALI. Protein immunoprecipitation indicated that TFEB and PGC-1α are interacting proteins. The luciferase reporter assay demonstrated that PGC-1α positively regulated TFEB in the LPS-induced primary type II alveolar epithelial cells. CONCLUSION PGC-1α protects LPS-induced ALI by decreasing inflammation and alleviating lung edema. The mechanism might be positive regulation of TFEB directly and then upregulation of mitophagy in LPS-induced ALI.
Collapse
Affiliation(s)
- Wei Liu
- Department of Respiratory, The Second Affiliated Hospital, The Air Force Military Medical University, Xi'an, 710038, ShaanXi, China
| | - Yanyan Li
- Department of Respiratory, The Second Affiliated Hospital, The Air Force Military Medical University, Xi'an, 710038, ShaanXi, China
| | - Liyan Bo
- Department of Respiratory, The Second Affiliated Hospital, The Air Force Military Medical University, Xi'an, 710038, ShaanXi, China
| | - Congcong Li
- Department of Respiratory, The General Hospital of Northern Theater, Shenyang, 110000, Liaoning, China
| | - Faguang Jin
- Department of Respiratory, The Second Affiliated Hospital, The Air Force Military Medical University, Xi'an, 710038, ShaanXi, China.
| |
Collapse
|
19
|
Alsharif KF, Almalki AA, Alsanie WF, Alzahrani KJ, Kabrah SM, Elshopakey GE, Alghamdi AAA, Lokman MS, Sberi HA, Bauomy AA, Albrakati A, Ramadan SS, Kassab RB, Abdel Moneim AE, Salem FEH. Protocatechuic acid attenuates lipopolysaccharide-induced septic lung injury in mice: The possible role through suppressing oxidative stress, inflammation and apoptosis. J Food Biochem 2021; 45:e13915. [PMID: 34472624 DOI: 10.1111/jfbc.13915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/14/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
Here, we investigated the protective efficacy of protocatechuic acid (PCA) against lipopolysaccharide (LPS)-induced septic lung injury. Eighty-two male Balb/c mice were divided into six groups: control, PCA30 (30 mg/kg), LPS (10 mg/kg), PCA10-LPS, PCA20-LPS, and PCA30-LPS treated with 10, 20 and 30 mg/kg PCA, respectively, for seven days before intraperitoneal LPS injection. PCA pre-treatment, especially at higher dose, significantly reduced LPS-induced lung tissue injury as indicated by increased heat shock protein 70 and antioxidant molecules (reduced glutathione, superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase) accompanied by lower oxidative stress indices (malondialdehyde and nitric oxide). PCA administration decreased inflammatory mediators including myeloperoxidase, nuclear factor kappa B (NF-κB p65), and pro-inflammatory cytokines, and prevented the development of apoptotic events in the lung tissue. At the molecular level, PCA downregulated mRNA expression of nitric oxide synthase 2, C/EBP homologous protein, and high mobility group box1 in the lungs of all PCA-LPS treated mice. Thus, PCA-pre-treatment effectively counteracted sepsis-induced acute lung injury in vivo by promoting and antioxidant status, while inhibiting inflammation and apoptosis. PRACTICAL IMPLICATIONS: Sepsis-mediated organ dysfunction and high mortality is aggravated by acute lung injury (ALI). Therefore, new therapeutic approaches are needed to encounter sepsis-mediated ALI. Protocatechuic acid (PCA) is a naturally occurring phenolic acid with various biological and pharmacological activities. PCA is abundant in edible plants including Allium cepa L., Oryza sativa L., Hibiscus sabdariffa, Prunus domestica L., and Eucommia ulmoides. In this investigation we studied the potential protective role of pure PCA (10, 20 and 30 mg/kg) on LPS-mediated septic lung injury in mice through examining oxidative challenge, inflammatory response, apoptotic events and histopathological changes in addition to evaluating the levels and mRNA expression of heat shock protein 70, C/EBP homologous protein and high mobility group box1 in the lung tissue. The recorded results showed that PCA pre-administration was able to significantly abrogate the damages in the lung tissue associated septic response. This protective effect comes from its strong antioxidant, anti-inflammatory, and anti-apoptotic activities, suggesting that PCA may be applied to alleviate ALI associated with the development of sepsis.
Collapse
Affiliation(s)
- Khalaf F Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Abdulraheem Ali Almalki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Walaa F Alsanie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Khalid J Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Saeed M Kabrah
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm AlQura University, Mecca, Saudi Arabia
| | - Gehad E Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | | | - Maha S Lokman
- Biology Department, College of Science and Humanities, Prince Sattam Bin Abdul Aziz University, Alkharj, Saudi Arabia.,Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Hassan Al Sberi
- Basic Medical Science, Histopathology Department, National Organization for Drug Control and Research, Giza, Egypt.,Department of Biology, Faculty of Science, Taif University, Taif, Saudi Arabia
| | - Amira A Bauomy
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt.,Department of Science Laboratories, College of Science and Arts, Qassim University, ArRass, Saudi Arabia
| | - Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Shimaa S Ramadan
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, Egypt
| | - Rami B Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt.,Department of Biology, Faculty of Science and Arts, Al Baha University, Almakhwah, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Fatma Elzahraa H Salem
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
20
|
Wang W, Xu N, Yu X, Zuo F, Liu J, Wang Y, Zhu G, Jia N, Ren H, Zhang J. Changes of Extravascular Lung Water as an Independent Prognostic Factor for Early Developed ARDS in Severely Burned Patients. J Burn Care Res 2021; 41:402-408. [PMID: 31720689 DOI: 10.1093/jbcr/irz189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
An important feature of acute respiratory distress syndrome (ARDS) is fluid lost into the interstitium of lung combined with its compromised reabsorption, resulting in the elevation of extravascular lung water (EVLW). Although ARDS is known as an early, common, and life-threatening complication in major burns, the issue of whether or how the EVLW index (EVLWI) correlates with its prognosis has not been identified yet. In this retrospectively study, 121 severely burned adults with ARDS occurred in 2 weeks postburn were analyzed and divided into two groups: survivors (73 patients) and nonsurvivors (48 patients) according to the 28-day outcome after injury. Compared with nonsurvivors, survivors exhibited bigger EVLWI reduction in day 2 after ARDS onset (ΔEVLWI2), with no differences in ARDS timing and other EVLWI variables. ΔEVLWI2, rather than EVLWI on 2 days after ARDS onset, was identified as an independent prognostic factor even after adjusting other significant factors by Cox proportional hazard analysis. ROC curve analysis showed that ΔEVLWI2 [AUC = 0.723, 95% CI = (0.631-0.816), P < .001] was a relative predictor for survival on 28-day postburn, with a threshold of 1.9 ml/kg (63.0% sensitivity, 77.1% specificity). Kaplan-Meier survival curve analysis confirmed a significantly higher survival rate on 28-day postburn in patients with ΔEVLWI2 > 1.9 ml/kg (log-rank test: χ 2 =14.780, P < .001). Taken together, our study demonstrated that ΔEVLWI2 is an independent prognostic factor for early ARDS in severe burns. ΔEVLWI2 higher than 1.9 ml/kg might predict a higher survival rate in those patients.
Collapse
Affiliation(s)
- Weiyi Wang
- Dalian Rehabilitation Recuperation Center of PLA Joint Logistics Support Force, Dalian, China.,School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ning Xu
- Department of Pathology, No. 984 Hospital of PLA, Beijing, China
| | - Xiaofeng Yu
- School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengli Zuo
- School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Liu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuan Wang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Guoqin Zhu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Naixin Jia
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Ren
- School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiaping Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
21
|
Ali FEM, Ahmed SF, Eltrawy AH, Yousef RS, Ali HS, Mahmoud AR, Abd-Elhamid TH. Pretreatment with Coenzyme Q10 Combined with Aescin Protects against Sepsis-Induced Acute Lung Injury. Cells Tissues Organs 2021; 210:195-217. [PMID: 34280918 DOI: 10.1159/000516192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/26/2021] [Indexed: 11/19/2022] Open
Abstract
Sepsis-associated acute lung injury (ALI) is a critical condition characterized by severe inflammatory response and mitochondrial dysfunction. Coenzyme Q10 (CoQ10) and aescin (AES) are well-known for their anti-inflammatory activities. However, their effects on lipopolysaccharide (LPS)-induced lung injury have not been explored yet. Here, we asked whether combined pretreatment with CoQ10 and AES synergistically prevents LPS-induced lung injury. Fifty male rats were randomized into 5 groups: (1) control; (2) LPS-treated, rats received a single i.p. injection of LPS (8 mg/kg); (3) CoQ10-pretreated, (4) AES-pretreated, or (5) combined-pretreated; animals received CoQ10 (100 mg/kg), AES (5 mg/kg), or both orally for 7 days before LPS injection. Combined CoQ10 and AES pretreatment significantly reduced lung injury markers; 52.42% reduction in serum C-reactive protein (CRP), 53.69% in alkaline phosphatase (ALKP) and 60.26% in lactate dehydrogenase (LDH) activities versus 44.58, 37.38, and 48.6% in CoQ10 and 33.81, 34.43, and 39.29% in AES-pretreated groups, respectively. Meanwhile, combination therapy significantly reduced interleukin (IL)-1β and tumor necrosis factor (TNF)-α expressions compared to monotherapy (p < 0.05). Additionally, combination therapy prevented LPS-induced histological and mitochondrial abnormalities greater than separate drugs. Western blotting indicated that combination therapy significantly suppressed nucleotide-binding oligomerization domain (NOD)-like receptors-3 (NLRP-3) inflammasome compared to separate drugs (p < 0.05). Further, combination therapy significantly decreased the expression of signaling cascades, p38 mitogen-activated protein kinases (p38 MAPK), nuclear factor kappa B (NF-κB)-p65, and extracellular-regulated kinases 1/2 (ERK1/2) versus monotherapy (p < 0.05). Interestingly, combined pretreatment significantly downregulated high mobility group box-1 (HMGB1) by 72.93%, and toll-like receptor 4 (TLR4) by -0.93-fold versus 61.92%, -0.83-fold in CoQ10 and 38.67%, -0.70-fold in AES pretreatment, respectively. Our results showed for the first time that the enhanced anti-inflammatory effect of combined CoQ10 and AES pretreatment prevented LPS-induced ALI via suppression of NLRP-3 inflammasome through regulation of HMGB1/TLR4 signaling pathway and mitochondrial stabilization.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Salwa F Ahmed
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amira H Eltrawy
- Department of Anatomy and Embryology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Reda S Yousef
- Department of Biochemistry, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Howaida S Ali
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Amany R Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Basic Medical Sciences, Unaizah College of Medicine and Medical Sciences, Qassim University, Unaizah, Saudi Arabia
| | - Tarek H Abd-Elhamid
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
22
|
Wu H, Hong X, Qu Y, Liu Z, Zhao Z, Liu C, Ji Q, Wang J, Xueli Q, Jianwei S, Cheng D, Feng ZC, Yuan S. The value of oxygen index and base excess in predicting the outcome of neonatal acute respiratory distress syndrome. J Pediatr (Rio J) 2021; 97:409-413. [PMID: 32822669 PMCID: PMC8166491 DOI: 10.1016/j.jped.2020.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE This study aimed to identify the predictors and threshold of failure in neonatal acute respiratory distress syndrome. METHODS Newborns with severe acute respiratory distress syndrome aged 0-28 days and gestational age ≥36 weeks were included in the study if their cases were managed with non-extra corporal membrane oxygenation treatments. Patients were divided into two groups according to whether they died before discharge. Predictors of non-extra corporal membrane oxygenation treatment failure were sought, and the threshold of predictors was calculated. RESULTS A total of 103 patients were included in the study. A total of 77 (74.8%) survived hospitalization and were discharged, whereas 26 (25.2%) died. Receiver operating characteristic analysis of oxygen index, pH, base excess, and combinations of these indicators demonstrated the advantage of the combination of oxygen index and base excess over the others variables regarding their predictive ability. The area under the curve for the combination of oxygen index and base excess was 0.865. When the cut-off values of oxygen index and base excess were 30.0 and -7.4, respectively, the sensitivity and specificity for predicting death were 77.0% and 84.0%, respectively. The model with base excess added a net reclassification improvement of 0.090 to the model without base excess. CONCLUSION The combination of oxygen index and base excess can be used as a predictor of outcomes in neonates receiving non-extra corporal membrane oxygenation treatment for acute respiratory distress syndrome. In neonates with acute respiratory distress syndrome, if oxygen index >30 and base excess <-7.4, non-extra corporal membrane oxygenation therapy is likely to lead to death.
Collapse
Affiliation(s)
- Hui Wu
- The First Hospital of Jilin University, Department of Neonatology, Changchun, China
| | - Xiaoyang Hong
- Affiliated Bayi Children's Hospital, Pediatric Intensive Care Unit, The Seventh Medical Center, PLA General Hospital, Beijing, China
| | - Yangming Qu
- The First Hospital of Jilin University, Department of Neonatology, Changchun, China
| | - Zhenqiu Liu
- Children's Hospital, Chongqing Medical University, Department of Neonatology, Chongqing, China
| | - Zhe Zhao
- Affiliated Bayi Children's Hospital, Pediatric Intensive Care Unit, The Seventh Medical Center, PLA General Hospital, Beijing, China
| | - Change Liu
- Affiliated Bayi Children's Hospital, Pediatric Intensive Care Unit, The Seventh Medical Center, PLA General Hospital, Beijing, China
| | - Qiong Ji
- The First Hospital of Jilin University, Department of Neonatology, Changchun, China
| | - Jie Wang
- Children's Hospital Affiliated of Zhengzhou University, Surgical Pediatric Intensive Care Unit, Henan, China
| | - Quan Xueli
- Children's Hospital Affiliated of Zhengzhou University, Surgical Pediatric Intensive Care Unit, Henan, China
| | - Sun Jianwei
- Henan Provincial People's Hospital, Department of Neonatology, Henan, China
| | - Dongliang Cheng
- Henan Provincial People's Hospital, Department of Neonatology, Henan, China
| | - Zhi-Chun Feng
- Affiliated Bayi Children's Hospital, Pediatric Intensive Care Unit, The Seventh Medical Center, PLA General Hospital, Beijing, China.
| | - Shi Yuan
- Children's Hospital, Chongqing Medical University, Department of Neonatology, Chongqing, China.
| |
Collapse
|
23
|
Louis G, Belveyre T, Jacquot A, Hochard H, Aissa N, Kimmoun A, Goetz C, Levy B, Novy E. Infection related catheter complications in patients undergoing prone positioning for acute respiratory distress syndrome: an exposed/unexposed study. BMC Infect Dis 2021; 21:534. [PMID: 34098888 PMCID: PMC8182343 DOI: 10.1186/s12879-021-06197-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Background Prone positioning (PP) is a standard of care for patients with moderate–severe acute respiratory distress syndrome (ARDS). While adverse events associated with PP are well-documented in the literature, research examining the effect of PP on the risk of infectious complications of intravascular catheters is lacking. Method All consecutive ARDS patients treated with PP were recruited retrospectively over a two-year period and formed the exposed group. Intensive care unit (ICU) patients during the same period without ARDS for whom PP was not conducted but who had an equivalent disease severity were matched 1:1 to the exposed group based on age, sex, centre, length of ICU stay and SAPS II (unexposed group). Infection-related catheter complications were defined by a composite criterion, including catheter tip colonization or intravascular catheter-related infection. Results A total of 101 exposed patients were included in the study. Most had direct ARDS (pneumonia). The median [Q1–Q3] PP session number was 2 [1–4]. These patients were matched with 101 unexposed patients. The mortality rates of the exposed and unexposed groups were 31 and 30%, respectively. The incidence of the composite criterion was 14.2/1000 in the exposed group compared with 8.2/1000 days in the control group (p = 0.09). Multivariate analysis identified PP as a factor related to catheter colonization or infection (p = 0.04). Conclusion Our data suggest that PP is associated with a higher risk of CVC infectious complications.
Collapse
Affiliation(s)
- Guillaume Louis
- Intensive Care Unit, Metz-Thionville Regional Hospital, Mercy Hospital, 1 allée de Château, 57085, Metz, France.
| | - Thibaut Belveyre
- Intensive Care Unit, Metz-Thionville Regional Hospital, Mercy Hospital, 1 allée de Château, 57085, Metz, France
| | - Audrey Jacquot
- Medical intensive Care Unit, University Hospital of Nancy, Brabois, France
| | - Hélène Hochard
- Department of Bacteriology, Metz-Thionville Regional Hospital, Mercy Hospital, Metz, France
| | - Nejla Aissa
- Department of Bacteriology, University Hospital of Nancy, Nancy, France
| | - Antoine Kimmoun
- Medical intensive Care Unit, University Hospital of Nancy, Brabois, France
| | - Christophe Goetz
- Clinical Research Support Unit, Metz-Thionville Regional Hospital, Metz, France
| | - Bruno Levy
- Medical intensive Care Unit, University Hospital of Nancy, Brabois, France
| | - Emmanuel Novy
- Intensive Care Unit, Metz-Thionville Regional Hospital, Mercy Hospital, 1 allée de Château, 57085, Metz, France
| |
Collapse
|
24
|
Surolia R, Li FJ, Wang Z, Kashyap M, Srivastava RK, Traylor AM, Singh P, Dsouza KG, Kim H, Pittet JF, Zmijewski JW, Agarwal A, Athar M, Ahmad A, Antony VB. NETosis in the pathogenesis of acute lung injury following cutaneous chemical burns. JCI Insight 2021; 6:147564. [PMID: 34027893 PMCID: PMC8262367 DOI: 10.1172/jci.insight.147564] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
Despite the high morbidity and mortality among patients with extensive cutaneous burns in the intensive care unit due to the development of acute respiratory distress syndrome, effective therapeutics remain to be determined. This is primarily because the mechanisms leading to acute lung injury (ALI) in these patients remain unknown. We test the hypothesis that cutaneous chemical burns promote lung injury due to systemic activation of neutrophils, in particular, toxicity mediated by the deployment of neutrophil extracellular traps (NETs). We also demonstrate the potential benefit of a peptidyl arginine deiminase 4 (PAD4) inhibitor to prevent NETosis and to preserve microvascular endothelial barrier function, thus reducing the severity of ALI in mice. Our data demonstrated that phenylarsine oxide (PAO) treatment of neutrophils caused increased intracellular Ca2+-associated PAD4 activity. A dermal chemical burn by lewisite or PAO resulted in PAD4 activation, NETosis, and ALI. NETs disrupted the barrier function of endothelial cells in human lung microvascular endothelial cell spheroids. Citrullinated histone 3 alone caused ALI in mice. Pharmacologic or genetic abrogation of PAD4 inhibited lung injury following cutaneous chemical burns. Cutaneous burns by lewisite and PAO caused ALI by PAD4-mediated NETosis. PAD4 inhibitors may have potential as countermeasures to suppress detrimental lung injury after chemical burns.
Collapse
Affiliation(s)
- Ranu Surolia
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine
| | - Fu Jun Li
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine
| | - Zheng Wang
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine
| | | | | | | | - Pooja Singh
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine
| | - Kevin G Dsouza
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine
| | | | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Anupam Agarwal
- Division of Nephrology, Department of Medicine.,Department of Veterans Affairs, Birmingham, Alabama, USA
| | | | - Aftab Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Veena B Antony
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine
| |
Collapse
|
25
|
Kasotakis G, Stanfield B, Haines K, Vatsaas C, Alger A, Vaslef SN, Brooks K, Agarwal S. Acute Respiratory Distress Syndrome (ARDS) after trauma: Improving incidence, but increasing mortality. J Crit Care 2021; 64:213-218. [PMID: 34022661 DOI: 10.1016/j.jcrc.2021.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 01/24/2023]
Abstract
PURPOSE Acute Respiratory Distress Syndrome (ARDS) is an infrequent, yet morbid inflammatory complication in injury victims. With the current project we sought to estimate trends in incidence, determine outcomes, and identify risk factors for ARDS and related mortality. MATERIALS & METHODS The national Trauma Quality Improvement Program dataset (2010-2014) was queried. Demographics, injury characteristics and outcomes were compared between patients who developed ARDS and those who did not. Logistic regression models were fitted for the development of ARDS and mortality respectively, adjusting for relevant confounders. RESULTS In the studied 808,195 TQIP patients, incidence of ARDS decreased over the study years (3-1.1%, p < 0.001), but related mortality increased (18.-21%, p = 0.001). ARDS patients spent an additional 14.7 ± 10.3 days in the hospital, 9.7 ± 7.9 in the ICU, and 6.6 ± 9.4 on mechanical ventilation (all p < 0.001). Older age, male gender, African American race increased risk for ARDS. Age, male gender, lower GCS and higher ISS also increased mortality risk among ARDS patients. Several pre-existing comorbidities including chronic alcohol use, diabetes, smoking, and respiratory disease also increased risk. CONCLUSION Although the incidence of ARDS after trauma appears to be declining, mortality is on the rise.
Collapse
Affiliation(s)
- George Kasotakis
- Department of Surgery, Duke University School of Medicine, United States of America.
| | - Brent Stanfield
- Department of Surgery, Duke University School of Medicine, United States of America.
| | - Krista Haines
- Department of Surgery, Duke University School of Medicine, United States of America.
| | - Cory Vatsaas
- Department of Surgery, Duke University School of Medicine, United States of America.
| | - Amy Alger
- Department of Surgery, Duke University School of Medicine, United States of America.
| | - Steven N Vaslef
- Department of Surgery, Duke University School of Medicine, United States of America.
| | - Kelli Brooks
- Department of Surgery, Duke University School of Medicine, United States of America.
| | - Suresh Agarwal
- Department of Surgery, Duke University School of Medicine, United States of America.
| |
Collapse
|
26
|
Probabilistic Dual-Hesitant Pythagorean Fuzzy Sets and Their Application in Multi-attribute Group Decision-Making. Cognit Comput 2021. [DOI: 10.1007/s12559-021-09858-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Tan W, Xu DY, Xu MJ, Wang ZF, Dai B, Li LL, Zhao HW, Wang W, Kang J. The efficacy and tolerance of prone positioning in non-intubation patients with acute hypoxemic respiratory failure and ARDS: a meta-analysis. Ther Adv Respir Dis 2021; 15:17534666211009407. [PMID: 33888007 PMCID: PMC8071979 DOI: 10.1177/17534666211009407] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background and aims: The application of prone positioning with acute hypoxemic respiratory failure (AHRF) or acute respiratory distress syndrome (ARDS) in non-intubation patients is increasing gradually, applying prone positioning for more high-flow nasal oxygen therapy (HFNC) and non-invasive ventilation (NIV) patients. This meta-analysis evaluates the efficacy and tolerance of prone positioning combined with non-invasive respiratory support in patients with AHRF or ARDS. Methods: We searched randomized controlled trials (RCTs) (prospective or retrospective cohort studies, RCTs and case series) published in PubMed, EMBASE and the Cochrane Central Register of Controlled Trials from 1 January 2000 to 1 July 2020. We included studies that compared prone and supine positioning with non-invasive respiratory support in awake patients with AHRF or ARDS. The meta-analyses used random effects models. The methodological quality of the RCTs was evaluated using the Newcastle–Ottawa quality assessment scale. Results: A total of 16 studies fulfilled selection criteria and included 243 patients. The aggregated intubation rate and mortality rate were 33% [95% confidence interval (CI): 0.26–0.42, I2 = 25%], 4% (95% CI: 0.01–0.07, I2 = 0%), respectively, and the intolerance rate was 7% (95% CI: 0.01–0.12, I2 = 5%). Prone positioning increased PaO2/FiO2 [mean difference (MD) = 47.89, 95% CI: 28.12–67.66; p < 0.00001, I2 = 67%] and SpO2 (MD = 4.58, 95% CI: 1.35–7.80, p = 0.005, I2 = 97%), whereas it reduced respiratory rate (MD = −5.01, 95% CI: −8.49 to −1.52, p = 0.005, I2 = 85%). Subgroup analyses demonstrated that the intubation rate of shorter duration prone (⩽5 h/day) and longer duration prone (>5 h/day) were 34% and 21%, respectively; and the mortality rate of shorter duration prone (⩽5 h/day) and longer duration prone (>5 h/day) were 6% and 0%, respectively. PaO2/FiO2 and SpO2 were significantly improved in COVID-19 patients and non-COVID-19 patients. Conclusion: Prone positioning could improve the oxygenation and reduce respiratory rate in both COVID-19 patients and non-COVID-19 patients with non-intubated AHRF or ARDS. The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Wei Tan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dong-Yang Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Meng-Jiao Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zan-Feng Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bing Dai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, Liaoning 110001, China
| | - Li-Li Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, Liaoning 110001, China
| | - Hong-Wen Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jian Kang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
28
|
The effect of preemptive airway pressure release ventilation on patients with high risk for acute respiratory distress syndrome: a randomized controlled trial. Braz J Anesthesiol 2021; 72:29-36. [PMID: 33905798 PMCID: PMC9373213 DOI: 10.1016/j.bjane.2021.03.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 03/14/2021] [Accepted: 03/19/2021] [Indexed: 11/23/2022] Open
Abstract
Background and objectives The objective of this study was to investigate the use of early APRV mode as a lung protective strategy compared to conventional methods with regard to ARDS development. Methods The study was designed as a randomized, non-blinded, single-center, superiority trial with two parallel groups and a primary endpoint of ARDS development. Patients under invasive mechanical ventilation who were not diagnosed with ARDS and had Lung Injury Prediction Score greater than 7 were included in the study. The patients were assigned to APRV and P-SIMV + PS mode groups. Results Patients were treated with P-SIMV+PS or APRV mode; 33 (50.8%) and 32 (49.2%), respectively. The P/F ratio values were higher in the APRV group on day 3 (p = 0.032). The fraction of inspired oxygen value was lower in the APRV group at day 7 (p = 0.011).While 5 of the 33 patients (15.2%) in the P-SIMV+PS group developed ARDS, one out of the 32 patients (3.1%) in the APRV group developed ARDS during follow-up (p = 0.197). The groups didn’t differ in terms of vasopressor/inotrope requirement, successful extubation rates, and/or mortality rates (p = 1.000, p = 0.911, p = 0.705, respectively). Duration of intensive care unit stay was 8 (2–11) days in the APRV group and 13 (8–81) days in the P-SIMV+PS group (p = 0.019). Conclusions The APRV mode can be used safely in selected groups of surgical and medical patients while preserving spontaneous respiration to a make benefit of its lung-protective effects. In comparison to the conventional mode, it is associated with improved oxygenation, higher mean airway pressures, and shorter intensive care unit stay. However, it does not reduce the sedation requirement, ARDS development, or mortality.
Collapse
|
29
|
Scott TE, Johnston AM, Keene DD, Rana M, Mahoney PF. Primary Blast Lung Injury: The UK Military Experience. Mil Med 2021; 185:e568-e572. [PMID: 31875895 DOI: 10.1093/milmed/usz453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION Primary blast lung injury occurs when an explosive shock wave passes through the thorax and transits through tissues of varying densities. It requires close proximity to an explosion and presents quick with respiratory distress in survivors. MATERIALS AND METHODS The Joint Theatre Trauma Registry and the Defence Statistics (Health) Database were interrogated for casualties injured as a result of an explosion during the conflict in Afghanistan. The case notes and imaging of casualties meeting the criteria for diagnosis were reviewed. Demographic and clinical data on casualties with primary blast lung injury were analyzed. RESULTS 848 blast-exposed casualties survived to discharge from intensive care, and 238 blast-exposed casualties were killed in action. Following exclusions, 111 case notes and all postmortem reports were reviewed in detail. About, 25 casualties had isolated primary blast lung injury (2.9% of casualties surviving to discharge from intensive care) and 31 nonsurvivors (13% of nonsurvivors) had the disease documented at postmortem. Severe cases of primary blast lung injury required an estimated average of 4.5 days of conventional mechanical ventilation. CONCLUSIONS 8.1% of blast exposed casualties suffered primary blast lung injury. It was a less severe disease than other nontraumatic forms of acute lung injury and did not cause deaths once a casualty had reached a combat support hospital. It was well managed with a relatively brief period of conventional mechanical ventilation.
Collapse
Affiliation(s)
- Timothy E Scott
- Academic Department of Military Anaesthesia and Critical Care, Royal Centre for Defence Medicine, ICT Centre, Birmingham B15 2SQ, UK
| | - Andrew M Johnston
- Academic Department of Military Anaesthesia and Critical Care, Royal Centre for Defence Medicine, ICT Centre, Birmingham B15 2SQ, UK
| | - Damian D Keene
- Academic Department of Military Anaesthesia and Critical Care, Royal Centre for Defence Medicine, ICT Centre, Birmingham B15 2SQ, UK
| | - Meenal Rana
- Department of Anaesthesia, Glenfield Hospital, Groby Road, Leicester, LE3 9QP UK
| | - Peter F Mahoney
- Academic Department of Military Anaesthesia and Critical Care, Royal Centre for Defence Medicine, ICT Centre, Birmingham B15 2SQ, UK
| |
Collapse
|
30
|
Zhang H, Mao YF, Zhao Y, Xu DF, Wang Y, Xu CF, Dong WW, Zhu XY, Ding N, Jiang L, Liu YJ. Upregulation of Matrix Metalloproteinase-9 Protects against Sepsis-Induced Acute Lung Injury via Promoting the Release of Soluble Receptor for Advanced Glycation End Products. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8889313. [PMID: 33628393 PMCID: PMC7889353 DOI: 10.1155/2021/8889313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/21/2020] [Accepted: 01/17/2021] [Indexed: 02/06/2023]
Abstract
Dysregulation of matrix metalloproteinase- (MMP-) 9 is implicated in the pathogenesis of acute lung injury (ALI). However, it remains controversial whether MMP-9 improves or deteriorates acute lung injury of different etiologies. The receptor for advanced glycation end products (RAGE) plays a critical role in the pathogenesis of acute lung injury. MMPs are known to mediate RAGE shedding and release of soluble RAGE (sRAGE), which can act as a decoy receptor by competitively inhibiting the binding of RAGE ligands to RAGE. Therefore, this study is aimed at clarifying whether and how pulmonary knockdown of MMP-9 affected sepsis-induced acute lung injury as well as the release of sRAGE in a murine cecal ligation and puncture (CLP) model. The analysis of GEO mouse sepsis datasets GSE15379, GSE52474, and GSE60088 revealed that the mRNA expression of MMP-9 was significantly upregulated in septic mouse lung tissues. Elevation of pulmonary MMP-9 mRNA and protein expressions was confirmed in CLP-induced mouse sepsis model. Intratracheal injection of MMP-9 siRNA resulted in an approximately 60% decrease in pulmonary MMP-9 expression. It was found that pulmonary knockdown of MMP-9 significantly increased mortality of sepsis and exacerbated sepsis-associated acute lung injury. Pulmonary MMP-9 knockdown also decreased sRAGE release and enhanced sepsis-induced activation of the RAGE/nuclear factor-κB (NF-κB) signaling pathway, meanwhile aggravating sepsis-induced oxidative stress and inflammation in lung tissues. In addition, administration of recombinant sRAGE protein suppressed the activation of the RAGE/NF-κB signaling pathway and ameliorated pulmonary oxidative stress, inflammation, and lung injury in CLP-induced septic mice. In conclusion, our data indicate that MMP-9-mediated RAGE shedding limits the severity of sepsis-associated pulmonary edema, inflammation, oxidative stress, and lung injury by suppressing the RAGE/NF-κB signaling pathway via the decoy receptor activities of sRAGE. MMP-9-mediated sRAGE production may serve as a self-limiting mechanism to control and resolve excessive inflammation and oxidative stress in the lung during sepsis.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yan-Fei Mao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Ying Zhao
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Dun-Feng Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Chu-Fan Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Wen-Wen Dong
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xiao-Yan Zhu
- Department of Physiology, Navy Medical University, Shanghai 200433, China
| | - Ning Ding
- Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yu-Jian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
31
|
Abstract
ABSTRACT Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are caused by an exaggerated inflammatory response arising from a wide variety of pulmonary and systemic insults. Lung tissue is composed of a variety of cell populations, including parenchymal and immune cells. Emerging evidence has revealed that multiple cell populations in the lung work in concert to regulate lung inflammation in response to both direct and indirect stimulations. To date, the question of how different types of pulmonary cells communicate with each other and subsequently regulate or modulate inflammatory cascades remains to be fully addressed. In this review, we provide an overview of current advancements in understanding the role of cell-cell interaction in the development of ALI and depict molecular mechanisms by which cell-cell interactions regulate lung inflammation, focusing on inter-cellular activities and signaling pathways that point to possible therapeutic opportunities for ALI/ARDS.
Collapse
Affiliation(s)
- Huiting Zhou
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Erica K. Fan
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Metz JK, Wiegand B, Schnur S, Knoth K, Schneider-Daum N, Groß H, Croston G, Reinheimer TM, Lehr CM, Hittinger M. Modulating the Barrier Function of Human Alveolar Epithelial (hAELVi) Cell Monolayers as a Model of Inflammation. Altern Lab Anim 2021; 48:252-267. [DOI: 10.1177/0261192920983015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The incidence of inflammatory lung diseases such as acute respiratory distress syndrome (ARDS) remains an important problem, particularly in the present time with the Covid-19 pandemic. However, an adequate in vitro test system to monitor the barrier function of the alveolar epithelium during inflammation and for assessing anti-inflammatory drugs is urgently needed. Therefore, we treated human Alveolar Epithelial Lentivirus-immortalised cells (hAELVi cells) with the pro-inflammatory cytokines TNF-α (25 ng/ml) and IFN-γ (30 ng/ml), in the presence or absence of hydrocortisone (HC). While TNF-α and IFN-γ are known to reduce epithelial barrier properties, HC could be expected to protect the barrier function and result in an anti-inflammatory effect. We investigated the impact of anti-inflammatory/inflammatory treatment on transepithelial electrical resistance (TEER) and the apparent permeability coefficient (P app) of the low permeability marker sodium fluorescein (NaFlu). After incubating hAELVi cells for 48 hours with a combination of TNF-α and IFN-γ, there was a significant decrease in TEER and a significant increase in the P app. The presence of HC maintained the TEER values and barrier properties, so that no significant P app change was observed. By using hAELVi cells to study anti-inflammatory drugs in vitro, the need for animal experiments could be reduced and pulmonary drug development accelerated.
Collapse
Affiliation(s)
- Julia Katharina Metz
- PharmBioTec GmbH, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | | | - Sabrina Schnur
- PharmBioTec GmbH, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | | | - Nicole Schneider-Daum
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | | | | | | | - Claus-Michael Lehr
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Marius Hittinger
- PharmBioTec GmbH, Saarbrücken, Germany
- 3RProducts Marius Hittinger, Blieskastel, Germany
| |
Collapse
|
33
|
Mikolka P, Curstedt T, Feinstein R, Larsson A, Grendar M, Rising A, Johansson J. Impact of synthetic surfactant CHF5633 with SP-B and SP-C analogues on lung function and inflammation in rabbit model of acute respiratory distress syndrome. Physiol Rep 2021; 9:e14700. [PMID: 33403805 PMCID: PMC7786196 DOI: 10.14814/phy2.14700] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/29/2020] [Accepted: 12/03/2020] [Indexed: 02/04/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with diffuse inflammation, alveolar epithelial damage, and leakage of plasma proteins into the alveolar space, which together contribute to inactivation of pulmonary surfactant and respiratory failure. Exogenous surfactant delivery is therefore considered to hold potential for ARDS treatment, but clinical trials with natural derived surfactant or synthetic surfactant containing a surfactant protein C (SP-C) analogue have been negative. Synthetic surfactant CHF5633, containing analogues of SP-B and SP-C, may be effective against ARDS. The aim here was to compare treatment effects of CHF5633 and animal-derived surfactant poractant alfa in animal model of ARDS. ARDS was induced in adult New Zealand rabbits by mild lung lavages followed by injurious ventilation until respiratory failure (P/F ratio <26.7 kPa). The animals were then treated with intratracheal bolus of 200 mg/kg CHF5633 or poractant alfa (Curosurf® ), or air as control. The animals were subsequently ventilated for an additional 4 hr and respiratory parameters were recorded regularly. Postmortem, histological analysis, degree of lung edema, and levels of the cytokines TNFα, IL-6, and IL-8 in lung homogenates were evaluated. Both surfactant preparations improved lung function, reduced the levels of pro-inflammatory cytokines, and degree of lung edema to very similar degrees versus the controls. No significant differences in any of the analyzed parameters were observed between the CHF5633- and poractant alfa-treated groups. This study indicates that single dose of CHF5633 improves lung function and attenuates inflammation as effectively as poractant alfa in experimental ARDS caused by injurious ventilation.
Collapse
Affiliation(s)
- Pavol Mikolka
- Division for NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetHuddingeSweden
- Biomedical Center MartinJessenius Faculty of Medicine in MartinComenius University in BratislavaMartinSlovakia
- Department of PhysiologyJessenius Faculty of Medicine in MartinComenius University in BratislavaMartinSlovakia
| | - Tore Curstedt
- Department of Molecular Medicine and SurgeryKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Riccardo Feinstein
- Department of PathologyThe Swedish National Veterinary InstituteUppsalaSweden
| | - Anders Larsson
- Hedenstierna LaboratoryDepartment of Surgical SciencesUppsala UniversityUppsalaSweden
| | - Marian Grendar
- Biomedical Center MartinJessenius Faculty of Medicine in MartinComenius University in BratislavaMartinSlovakia
| | - Anna Rising
- Division for NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetHuddingeSweden
- Department of Anatomy, Physiology and BiochemistrySwedish University of Agricultural SciencesUppsalaSweden
| | - Jan Johansson
- Division for NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetHuddingeSweden
| |
Collapse
|
34
|
Xue H, Li M. Protective effect of pterostilbene on sepsis-induced acute lung injury in a rat model via the JAK2/STAT3 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1452. [PMID: 33313197 PMCID: PMC7723647 DOI: 10.21037/atm-20-5814] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Bacterial infection is one of the most common causes of sepsis, with acute lung injury (ALI) being a related complication. Pterostilbene (PTS) is extracted from blueberries, peanuts, and grapes, and has numerous pharmacologic activities. The aim of the present study was to explore the underlying role of PTS protects against sepsis-mediated ALI. Methods We established a sepsis model induced by cecal ligation and puncture (CLP) in rats. The rats were randomly divided into five groups (n=5 each): sham group, CLP group, Dexmedetomidine group (Dex, 50 µg/kg) and PTS groups (25 and 50 mg/kg). Twenty-hours hours after CLP, PTS was intraperitoneally injected for 14 continuous days. The rats were killed, and blood and lung tissue were collected for pathological analysis and mRNA and protein detection. Results Our findings showed that PTS reduced the wet/dry ratio and ameliorated sepsis-induced pulmonary fibrosis (PF), which was associated with improvement of pathological damage in lung tissues. We also observed the inhibitory effect of PTS on apoptosis and release of inflammatory cytokines (i.e., tumor necrosis factor-α, interleukin-6, and monocyte chemotactic protein 1). In addition, PTS markedly suppressed the phosphorylation levels of Janus kinase-2 (JAK2) and signal transducer and activator of transcription 3 (STAT3). Conclusions Our results indicated that PTS inhibited the PF, apoptosis, and inflammatory response via the JAK2/STAT3 pathway in a sepsis-induced ALI rat model, providing a candidate for drug therapy of sepsis-induced ALI.
Collapse
Affiliation(s)
- Hua Xue
- Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
35
|
Nan CC, Zhang N, Cheung KCP, Zhang HD, Li W, Hong CY, Chen HS, Liu XY, Li N, Cheng L. Knockdown of lncRNA MALAT1 Alleviates LPS-Induced Acute Lung Injury via Inhibiting Apoptosis Through the miR-194-5p/FOXP2 Axis. Front Cell Dev Biol 2020; 8:586869. [PMID: 33117815 PMCID: PMC7575725 DOI: 10.3389/fcell.2020.586869] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/02/2020] [Indexed: 01/07/2023] Open
Abstract
Purpose We aimed to identify and verify the key genes and lncRNAs associated with acute lung injury (ALI) and explore the pathogenesis of ALI. Research showed that lower expression of the lncRNA metastasis-associated lung carcinoma transcript 1 (MALAT1) alleviates lung injury induced by lipopolysaccharide (LPS). Nevertheless, the mechanisms of MALAT1 on cellular apoptosis remain unclear in LPS-stimulated ALI. We investigated the mechanism of MALAT1 in modulating the apoptosis of LPS-induced human pulmonary alveolar epithelial cells (HPAEpiC). Methods Differentially expressed lncRNAs between the ALI samples and normal controls were identified using gene expression profiles. ALI-related genes were determined by the overlap of differentially expressed genes (DEGs), genes correlated with lung, genes correlated with key lncRNAs, and genes sharing significantly high proportions of microRNA targets with MALAT1. Quantitative real-time PCR (qPCR) was applied to detect the expression of MALAT1, microRNA (miR)-194-5p, and forkhead box P2 (FOXP2) mRNA in 1 μg/ml LPS-treated HPAEpiC. MALAT1 knockdown vectors, miR-194-5p inhibitors, and ov-FOXP2 were constructed and used to transfect HPAEpiC. The influence of MALAT1 knockdown on LPS-induced HPAEpiC proliferation and apoptosis via the miR-194-5p/FOXP2 axis was determined using Cell counting kit-8 (CCK-8) assay, flow cytometry, and Western blotting analysis, respectively. The interactions between MALAT1, miR-194-5p, and FOXP2 were verified using dual-luciferase reporter gene assay. Results We identified a key lncRNA (MALAT1) and three key genes (EYA1, WNT5A, and FOXP2) that are closely correlated with the pathogenesis of ALI. LPS stimulation promoted MALAT1 expression and apoptosis and also inhibited HPAEpiC viability. MALAT1 knockdown significantly improved viability and suppressed the apoptosis of LPS-stimulated HPAEpiC. Moreover, MALAT1 directly targeted miR-194-5p, a downregulated miRNA in LPS-stimulated HPAEpiC, when FOXP2 was overexpressed. MALAT1 knockdown led to the overexpression of miR-194-5p and restrained FOXP2 expression. Furthermore, inhibition of miR-194-5p exerted a rescue effect on MALAT1 knockdown of FOXP2, whereas the overexpression of FOXP2 reversed the effect of MALAT1 knockdown on viability and apoptosis of LPS-stimulated HPAEpiC. Conclusion Our results demonstrated that MALAT1 knockdown alleviated HPAEpiC apoptosis by competitively binding to miR-194-5p and then elevating the inhibitory effect on its target FOXP2. These data provide a novel insight into the role of MALAT1 in the progression of ALI and potential diagnostic and therapeutic strategies for ALI patients.
Collapse
Affiliation(s)
- Chuan-Chuan Nan
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Ning Zhang
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China.,Department of Stomatology Center, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Kenneth C P Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, China
| | - Hua-Dong Zhang
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Wei Li
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Cheng-Ying Hong
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Huai-Sheng Chen
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Xue-Yan Liu
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Nan Li
- Department of Stomatology Center, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Lixin Cheng
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
36
|
Hu Q, Wang Q, Han C, Yang Y. Sufentanil attenuates inflammation and oxidative stress in sepsis-induced acute lung injury by downregulating KNG1 expression. Mol Med Rep 2020; 22:4298-4306. [PMID: 33000200 PMCID: PMC7533471 DOI: 10.3892/mmr.2020.11526] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to investigate the effects of sufentanil on sepsis-induced acute lung injury (ALI), and identify the potential molecular mechanisms underlying its effect. In order to achieve this, a rat sepsis model was established. Following treatment with sufentanil, the lung wet/dry (W/D) weight ratio was calculated. Histopathological analysis was performed via hematoxylin and eosin staining. Levels of inflammatory factors in bronchoalveolar lavage fluid were determined via ELISA. Furthermore, malondialdehyde (MDA) content and the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) in tissue homogenates were assessed using commercial kits. Western blot analysis was performed to determine kininogen-1 (KNG1) protein expression. In addition, alveolar epithelial type II cells (AEC II) were stimulated with lipopolysaccharide (LPS) to mimic ALI. The levels of inflammation and oxidative stress were evaluated following overexpression of KNG1. Protein expression levels of nuclear factor-κB (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling were determined via western blot analysis. The results of the present study demonstrated that sufentanil alleviated histopathological injury and the W/D ratio in lung tissue. Following treatment with sufentanil, levels of inflammatory factors also decreased, accompanied by decreased concentrations of MDA, and increased activities of SOD, CAT and GSH-Px. Notably, KNG1 was decreased in lung tissues following treatment with sufentanil. Furthermore, overexpression of KNG1 attenuated the inhibitory effects of sufentanil on LPS-induced inflammation and oxidative stress in AEC II. Sufentanil markedly downregulated NF-κB expression, while upregulating Nrf2 and HO-1 expression levels, which was reversed following overexpression of KNG1. Taken together, the results of the present study suggested that sufentanil may alleviate inflammation and oxidative stress in sepsis-induced ALI by downregulating KNG1 expression.
Collapse
Affiliation(s)
- Quan Hu
- Intensive Care Unit, The First People's Hospital, Wuhan, Hubei 430200, P.R. China
| | - Qin Wang
- Department of Pathology, Hubei Women and Children Health Care Hospital, Wuhan, Hubei 430200, P.R. China
| | - Chuangang Han
- Department of Anesthesiology, The First People's Hospital, Wuhan, Hubei 430200, P.R. China
| | - Yan Yang
- Department of Anesthesiology, The First People's Hospital, Wuhan, Hubei 430200, P.R. China
| |
Collapse
|
37
|
Mikolka P, Kosutova P, Kolomaznik M, Topercerova J, Kopincova J, Calkovska A, Mokra D. Effect of different dosages of dexamethasone therapy on lung function and inflammation in an early phase of acute respiratory distress syndrome model. Physiol Res 2020; 68:S253-S263. [PMID: 31928043 DOI: 10.33549/physiolres.934364] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Inflammation associated with acute respiratory distress syndrome (ARDS) can damage the alveolar epithelium and surfactant and worsen the respiratory failure. Glucocorticoids (GC) appear to be a rational therapeutic approach, but the effect is still unclear, especially for early administration and low-dose. In this study we compared two low doses of dexamethasone in early phase of surfactant-depleted model of acute respiratory distress syndrome (ARDS). In the study, lung-lavaged New Zealand rabbits with respiratory failure (PaO(2)<26.7 kPa in FiO(2) 1.0) were treated with intravenous dexamethasone (DEX): 0.5 mg/kg (DEX-0.5) and 1.0 mg/kg (DEX-1.0), or were untreated (ARDS). Animals without ARDS served as controls. Respiratory parameters, lung edema, leukocyte shifts, markers of inflammation and oxidative damage in the plasma and lung were evaluated. Both doses of DEX improved the lung function vs. untreated animals. DEX-1.0 had faster onset with significant improvement in gas exchange and ventilation efficiency vs. DEX-0.5. DEX-1.0 showed a trend to reduce lung neutrophils, local oxidative damage, and levels of TNFalpha, IL-6, IL-8 more effectively than DEX-0.5 vs. ARDS group. Both dosages of dexamethasone significantly improved the lung function and suppressed inflammation in early phase ARDS, while some additional enhancement was observed for higher dose (1 mg/kg) of DEX.
Collapse
Affiliation(s)
- P Mikolka
- Department of Physiology and Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | | | | | | | | | | | |
Collapse
|
38
|
Gori E, Pierini A, Ceccherini G, Citi S, Mannucci T, Lippi I, Marchetti V. Pulmonary complications in dogs with acute presentation of pancreatitis. BMC Vet Res 2020; 16:209. [PMID: 32571307 PMCID: PMC7310026 DOI: 10.1186/s12917-020-02427-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 06/15/2020] [Indexed: 12/31/2022] Open
Abstract
Background In humans, respiratory complications in patients with acute pancreatitis (AP) are a common life-threatening comorbidity. Since possible lung impairment has not been individually evaluated in canine AP, the aims of the present study were to: (1) describe the prevalence, types and severity of pulmonary complications in dogs with acute presentation of AP, and (2) evaluate their association with mortality. AP diagnosis was based on compatible clinical and laboratory parameters, abnormal canine pancreatic-lipase test, and positive abdominal ultrasound within 48 h from admission. The canine acute pancreatitis severity score (CAPS) was calculated for each dog at admission. Arterial blood gas analysis and thoracic radiography were performed at admission. Thoracic radiography was classified on the basis of pulmonary pattern (normal, interstitial or alveolar) and a modified lung injury score (mLIS) was applied to the ventrodorsal projections for each dog. VetALI/VetARDS were diagnosed using current veterinary consensus. Dogs were divided into non-survivors or survivors (hospital discharge). Clinical, radiological and blood gas parameters collected at presentation were compared between survivors and non-survivors and associated with mortality. Results This prospective cohort study included twenty-six client-owned dogs with AP. Twelve out of twenty-six dogs (46%) died or were euthanized. At admission, thirteen dogs showed respiratory distress at physical examination, which was associated with death (P < 0.001). Radiographic abnormalities were found in twenty-one dogs: alveolar (n = 11) and interstitial pattern (n = 10). Radiographic alterations and mLIS score were both associated with death (P = 0.02 and P = 0.0023). The results of the arterial blood-gas evaluation showed that non-survivors had lower PaCO2 and HCO3− levels, and higher A-a gradient than survivors (P = 0.0014, P = 0.019 and P = 0.004, respectively). Specifically, three dogs had aspiration pneumonia, and VetALI was diagnosed in nine dogs (34.6%), and no dogs met the criteria for VetARDS. The presence of VetALI was associated with mortality (P < 0.001). Conclusions As with humans, possible lung impairments, such as VetALI, should be investigated in dogs with acute presentation of pancreatitis.
Collapse
Affiliation(s)
- Eleonora Gori
- Department of Veterinary Sciences, Veterinary Teaching Hospital "Mario Modenato", University of Pisa, Via Livornese Lato monte, San Piero a Grado, 56122, Pisa, Italy
| | - Alessio Pierini
- Department of Veterinary Sciences, Veterinary Teaching Hospital "Mario Modenato", University of Pisa, Via Livornese Lato monte, San Piero a Grado, 56122, Pisa, Italy.
| | - Gianila Ceccherini
- Department of Veterinary Sciences, Veterinary Teaching Hospital "Mario Modenato", University of Pisa, Via Livornese Lato monte, San Piero a Grado, 56122, Pisa, Italy
| | - Simonetta Citi
- Department of Veterinary Sciences, Veterinary Teaching Hospital "Mario Modenato", University of Pisa, Via Livornese Lato monte, San Piero a Grado, 56122, Pisa, Italy
| | - Tommaso Mannucci
- Department of Veterinary Sciences, Veterinary Teaching Hospital "Mario Modenato", University of Pisa, Via Livornese Lato monte, San Piero a Grado, 56122, Pisa, Italy
| | - Ilaria Lippi
- Department of Veterinary Sciences, Veterinary Teaching Hospital "Mario Modenato", University of Pisa, Via Livornese Lato monte, San Piero a Grado, 56122, Pisa, Italy
| | - Veronica Marchetti
- Department of Veterinary Sciences, Veterinary Teaching Hospital "Mario Modenato", University of Pisa, Via Livornese Lato monte, San Piero a Grado, 56122, Pisa, Italy
| |
Collapse
|
39
|
Prognostic Value of N-terminal Probrain Natriuretic Peptide for Patients with Acute Respiratory Distress Syndrome: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3472615. [PMID: 32337240 PMCID: PMC7165325 DOI: 10.1155/2020/3472615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 12/16/2019] [Accepted: 02/26/2020] [Indexed: 01/02/2023]
Abstract
Objectives The mortality rate of patients with acute respiratory distress syndrome (ARDS) is high. Hence, it is crucial to identify a reliable biomarker with wide clinical applications for predicting the prognosis of patients with ARDS. This systematic review and meta-analysis was conducted to investigate the value of plasma N-terminal probrain natriuretic peptide (NT-proBNP) for predicting mortality in patients with ARDS. Methods An electronic search of databases including PubMed, Web of Science, Cochrane Library, and Chinese National Knowledge Infrastructure was conducted up to May 31, 2019, without language restrictions. The quality of the included studies was evaluated using QUADAS-2. Data were extracted and analyzed to obtain pooled estimates of sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio. A forest graph was used to evaluate heterogeneity. Potential causes of heterogeneity were further explored by subgroup analysis based on the testing day, testing method, observation endpoint, or cut-off points. A summary receiver operating characteristic curve was drawn to obtain the pooled area under the curve. Results A total of 7 studies involving 581 patients with ARDS were included. The pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio were as follows: 0.79 (95% CI: 0.72–0.84), 0.79 (95% CI: 0.66–0.88), 3.68 (95% CI: 2.16–6.28), 0.27 (95% CI: 0.20–0.38), and 13.58 (95% CI: 6.17–29.90), respectively. The results of subgroup analysis showed that the testing day influenced the summary sensitivity and that the cut-off points influenced the summary sensitivity and specificity. Conclusion Our results indicate that elevated plasma NT-proBNP levels have a moderate value for predicting the mortality of patients with ARDS.
Collapse
|
40
|
Hu Y, Yang H, Ding X, Tao L, Liu M, Zhang C. A sesquiterpenoid from Ligularia pleurocaulis modulated macrophages polarisation through TLR4 pathway. Nat Prod Res 2020; 35:4853-4856. [PMID: 32233670 DOI: 10.1080/14786419.2020.1736068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
An eremophilane-type sesquiterpenoid (EPS), 3-oxo-eremophila-1,7(11)-dien-12,8β-olide, has been isolated from anti-inflammatory folk herbs, Ligularia pleurocaulis. The aim of present study is to explore protective effects of EPS on lipopolysaccharide (LPS)-induced inflammatory responses in acute lung injury (ALI). EPS treatments (40 and 80 mg/kg) significantly ameliorated LPS-stimulated pathological changes in lungs. Furthermore, in vivo and in vitro mechanism studies suggest that EPS exerts its protective effects on LPS-induced ALI by regulating macrophage polarisation via suppression of TLR4/MyD88-mediated MAPK and NF-κB signaling pathways, and EPS may be useful for the prevention on ALI in the clinical setting.
Collapse
Affiliation(s)
- Yang Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hua Yang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Xiaoqian Ding
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Lijun Tao
- School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Minyan Liu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Hebei Yiling Academy of Medical Limited Company, Shijiazhuang, People's Republic of China
| | - Chaofeng Zhang
- School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
41
|
Hu Y, Yang H, Ding X, Liu J, Wang X, Hu L, Liu M, Zhang C. Anti-inflammatory octahydroindolizine alkaloid enantiomers from Dendrobium crepidatum. Bioorg Chem 2020; 100:103809. [PMID: 32361293 DOI: 10.1016/j.bioorg.2020.103809] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/25/2020] [Accepted: 03/29/2020] [Indexed: 11/30/2022]
Abstract
Six pairs of octahydroindolizine-type alkaloid enantiomers (1-6) including three new compounds [(-)-1/(+)-1, 2] were isolated from the stems of Dendrobium crepidatum. Their structures including the absolute configurations were elucidated by extensive spectroscopic analyses and comparison between the experimental and calculated electronic circular dichroism (ECD). All compounds were examined for their inhibitory effects on nitric oxide (NO) production induced by lipopolysaccharide (LPS) in RAW264.7 cells. It was found that compounds (+)-1, 2 and (+)-6 exhibited pronounced inhibition on NO production with IC50 values in the range of 3.62-16.11 µM, being more active than the positive control, dexamethasone (IC50 = 47.04 µM). In vivo, compound 6 (100, 50 and 10 mg/kg) showed protective effects against LPS-induced acute lung injury (ALI) in mice.
Collapse
Affiliation(s)
- Yang Hu
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China; Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Hua Yang
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Xiaoqian Ding
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Jian Liu
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Xiachang Wang
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China.
| | - Minyan Liu
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China; Hebei Yiling Academy of Medical Limited Company, Shijiazhuang 050035, People's Republic of China.
| | - Chaofeng Zhang
- Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.
| |
Collapse
|
42
|
Shah J, Rana SS. Acute respiratory distress syndrome in acute pancreatitis. Indian J Gastroenterol 2020; 39:123-132. [PMID: 32285399 DOI: 10.1007/s12664-020-01016-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Development of organ failure is one of the major determinants of mortality in patients with acute pancreatitis (AP). Acute respiratory distress syndrome (ARDS) is an important cause of respiratory failure in AP and is associated with high mortality. Pathogenesis of ARDS in AP is incompletely understood. Release of various cytokines plays an important role in development of ARDS in AP. Increased gut permeability due to various toxins, inflammatory mediators, and pancreatic enzymes potentiates lung injury by gut-lymph-lung axis leading on to increased translocation of bacterial endotoxins. Various scoring systems, serum levels of various cytokines and lung ultrasound have been evaluated for prediction of development of ARDS in AP with varying results. Various drugs have shown encouraging results in prevention of ARDS in animal models but these encouraging results in animal models are yet to be confirmed in clinical studies. There is no specific effective treatment for ARDS. Treatment of sepsis and local complications of AP should be done according to the standard management strategies. Lung protective ventilatory strategies are of paramount importance to improve outcome of patients of AP with ARDS and therefore effective coordination between gastroenterologists and intensivists is needed for effective management of these patients.
Collapse
Affiliation(s)
- Jimil Shah
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160 012, India
| | - Surinder S Rana
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160 012, India.
| |
Collapse
|
43
|
Birkner DR, Halvachizadeh S, Pape HC, Pfeifer R. Mortality of Adult Respiratory Distress Syndrome in Trauma Patients: A Systematic Review over a Period of Four Decades. World J Surg 2020; 44:2243-2254. [DOI: 10.1007/s00268-020-05483-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
Xie Y, Barbosa ACS, Xu M, Oberly PJ, Ren S, Gibbs RB, Poloyac SM, Song WC, Fan J, Xie W. Hepatic Estrogen Sulfotransferase Distantly Sensitizes Mice to Hemorrhagic Shock-Induced Acute Lung Injury. Endocrinology 2020; 161:5677524. [PMID: 31837219 PMCID: PMC6970454 DOI: 10.1210/endocr/bqz031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/12/2019] [Indexed: 12/17/2022]
Abstract
Hemorrhagic shock (HS) is a potential life-threatening condition that may lead to injury to multiple organs, including the lung. The estrogen sulfotransferase (EST, or SULT1E1) is a conjugating enzyme that sulfonates and deactivates estrogens. In this report, we showed that the expression of Est was markedly induced in the liver but not in the lung of female mice subject to HS and resuscitation. Genetic ablation or pharmacological inhibition of Est effectively protected female mice from HS-induced acute lung injury (ALI), including interstitial edema, neutrophil mobilization and infiltration, and inflammation. The pulmonoprotective effect of Est ablation or inhibition was sex-specific, because the HS-induced ALI was not affected in male Est-/- mice. Mechanistically, the pulmonoprotective phenotype in female Est-/- mice was accompanied by increased lung and circulating levels of estrogens, attenuated pulmonary inflammation, and inhibition of neutrophil mobilization from the bone marrow and neutrophil infiltration to the lung, whereas the pulmonoprotective effect was abolished upon ovariectomy, suggesting that the protection was estrogen dependent. The pulmonoprotective effect of Est ablation was also tissue specific, as loss of Est had little effect on HS-induced liver injury. Moreover, transgenic reconstitution of human EST in the liver of global Est-/- mice abolished the pulmonoprotective effect, suggesting that it is the EST in the liver that sensitizes mice to HS-induced ALI. Taken together, our results revealed a sex- and tissue-specific role of EST in HS-induced ALI. Pharmacological inhibition of EST may represent an effective approach to manage HS-induced ALI.
Collapse
Affiliation(s)
- Yang Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - Anne Caroline S Barbosa
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - Patrick J Oberly
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - Songrong Ren
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - Robert B Gibbs
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - Samuel M Poloyac
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jie Fan
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
- Surgical Research, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Correspondence: Dr. Wen Xie, Center for Pharmacogenetics and Department of 17 Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261. E-mail:
| |
Collapse
|
45
|
Mechanistic target of rapamycin-mediated autophagy is involved in the alleviation of lipopolysaccharide-induced acute lung injury in rats. Int Immunopharmacol 2020; 78:105790. [DOI: 10.1016/j.intimp.2019.105790] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 01/01/2023]
|
46
|
Lam NN, Hung TD, Hung DK. Impact of “opening the lung” ventilatory strategy on burn patients with acute respiratory distress syndrome. Burns 2019; 45:1841-1847. [DOI: 10.1016/j.burns.2019.05.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
|
47
|
Mitochondrial Coenzyme Q Protects Sepsis-Induced Acute Lung Injury by Activating PI3K/Akt/GSK-3 β/mTOR Pathway in Rats. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5240898. [PMID: 31815144 PMCID: PMC6878790 DOI: 10.1155/2019/5240898] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/10/2019] [Accepted: 10/25/2019] [Indexed: 12/21/2022]
Abstract
The aim of our study was to assess the effects of mitochondrial coenzyme Q (MitoQ) on sepsis-induced acute lung injury (ALI) and investigate its possible mechanisms. The cecal ligation and puncture (CLP) method was used to establish a septic ALI model. Rats were randomly divided into Con group, CLP group, MitoQ group, and MitoQ + LY294002 group. The survival rate of the rats was recorded, and the survival rate curve was plotted. Moreover, the ratio of wet/dry weight (W/D) in lung tissue was measured. The activity of myeloperoxidase (MPO) was measured by using the MPO colorimetric activity assay kit. The levels of high-mobility group box 1 (HMGB1) and interleukin-6 (IL-6), macrophage inflammatory protein 2 (MIP2), and keratinocyte chemoattractant (KC) were analyzed by ELISA. The histopathological changes were measured by HE staining, and the lung injury was scored. TUNEL assay was applied to detect the apoptotic cells in lung tissue. The protein expressions were detected by western blot. MitoQ increased the survival rate and alleviated pulmonary edema in septic ALI rats. In addition, MitoQ inhibited the MPO activity and decreased the levels of HMGB1 and IL-6. After treatment with MitoQ, alveolar wall edema, inflammatory cell infiltration, and red blood cell exudation were relieved. MitoQ inhibited cell apoptosis in lung tissue of septic ALI rats. Meanwhile, MitoQ treatment remarkedly increased the expression of p-Akt, p-GSK-3β, and p-mTOR but decreased Bax, caspase-3, caspase-9, Beclin-1, and LC-3II/LC-3I. The effects of MitoQ were significantly reversed by the PI3K inhibitor (LY294002). Our study demonstrated that MitoQ could protect sepsis-induced acute lung injury by activating the PI3K/Akt/GSK-3β/mTOR pathway in rats.
Collapse
|
48
|
Zebialowicz Ahlström J, Massaro F, Mikolka P, Feinstein R, Perchiazzi G, Basabe-Burgos O, Curstedt T, Larsson A, Johansson J, Rising A. Synthetic surfactant with a recombinant surfactant protein C analogue improves lung function and attenuates inflammation in a model of acute respiratory distress syndrome in adult rabbits. Respir Res 2019; 20:245. [PMID: 31694668 PMCID: PMC6836435 DOI: 10.1186/s12931-019-1220-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023] Open
Abstract
AIM In acute respiratory distress syndrome (ARDS) damaged alveolar epithelium, leakage of plasma proteins into the alveolar space and inactivation of pulmonary surfactant lead to respiratory dysfunction. Lung function could potentially be restored with exogenous surfactant therapy, but clinical trials have so far been disappointing. These negative results may be explained by inactivation and/or too low doses of the administered surfactant. Surfactant based on a recombinant surfactant protein C analogue (rSP-C33Leu) is easy to produce and in this study we compared its effects on lung function and inflammation with a commercial surfactant preparation in an adult rabbit model of ARDS. METHODS ARDS was induced in adult New Zealand rabbits by mild lung-lavages followed by injurious ventilation (VT 20 m/kg body weight) until P/F ratio < 26.7 kPa. The animals were treated with two intratracheal boluses of 2.5 mL/kg of 2% rSP-C33Leu in DPPC/egg PC/POPG, 50:40:10 or poractant alfa (Curosurf®), both surfactants containing 80 mg phospholipids/mL, or air as control. The animals were subsequently ventilated (VT 8-9 m/kg body weight) for an additional 3 h and lung function parameters were recorded. Histological appearance of the lungs, degree of lung oedema and levels of the cytokines TNFα IL-6 and IL-8 in lung homogenates were evaluated. RESULTS Both surfactant preparations improved lung function vs. the control group and also reduced inflammation scores, production of pro-inflammatory cytokines, and formation of lung oedema to similar degrees. Poractant alfa improved compliance at 1 h, P/F ratio and PaO2 at 1.5 h compared to rSP-C33Leu surfactant. CONCLUSION This study indicates that treatment of experimental ARDS with synthetic lung surfactant based on rSP-C33Leu improves lung function and attenuates inflammation.
Collapse
Affiliation(s)
- J Zebialowicz Ahlström
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - F Massaro
- Anesthesia and Intesive Care, Villa Anthea Hospital, Bari, Italy
| | - P Mikolka
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - R Feinstein
- Department of Pathology, The Swedish National Veterinary Institute, Uppsala, Sweden
| | - G Perchiazzi
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - O Basabe-Burgos
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - T Curstedt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - A Larsson
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - J Johansson
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - A Rising
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden. .,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| |
Collapse
|
49
|
Partial liquid ventilation–induced mild hypothermia improves the lung function and alleviates the inflammatory response during acute respiratory distress syndrome in canines. Biomed Pharmacother 2019; 118:109344. [PMID: 31545246 PMCID: PMC9386951 DOI: 10.1016/j.biopha.2019.109344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/02/2019] [Accepted: 08/07/2019] [Indexed: 12/29/2022] Open
|
50
|
Veith AC, Bou Aram B, Jiang W, Wang L, Zhou G, Jefcoate CR, Couroucli XI, Lingappan K, Moorthy B. Mice Lacking the Cytochrome P450 1B1 Gene Are Less Susceptible to Hyperoxic Lung Injury Than Wild Type. Toxicol Sci 2019; 165:462-474. [PMID: 29939353 DOI: 10.1093/toxsci/kfy154] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Supplemental oxygen is a life-saving intervention administered to individuals suffering from respiratory distress, including adults with acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Despite the clinical benefit, supplemental oxygen can create a hyperoxic environment that increases reactive oxygen species, oxidative stress, and lung injury. We have previously shown that cytochrome P450 (CYP)1A enzymes decrease susceptibility to hyperoxia-induced lung injury. In this investigation, we determined the role of CYP1B1 in hyperoxic lung injury in vivo. Eight- to ten-week old C57BL/6 wild type (WT) and Cyp1b1-/- mice were exposed to hyperoxia (>95% O2) for 24-72 h or maintained in room air (21% O2). Lung injury was assessed by histology and lung weight to body weight (LW/BW) ratios. Extent of inflammation was determined by assessing pulmonary neutrophil infiltration and cytokine levels. Lipid peroxidation markers were quantified by gas chromatography mass spectrometry, and oxidative DNA adducts were quantified by 32P-postlabeling as markers of oxidative stress. We found that Cyp1b1-/- mice displayed attenuation of lung weight and pulmonary edema, particularly after 48-72 h of hyperoxia compared with WT controls. Further, Cyp1b1-/- mice displayed decreased levels of pulmonary oxidative DNA adducts and pulmonary isofurans after 24 h of hyperoxia. Cyp1b1-/- mice also showed increased pulmonary CYP1A1 and 1A2 and mRNA expression. In summary, our results support the hypothesis that Cyp1b1-/- mice display decreased hyperoxic lung injury than wild type counterparts and that CYP1B1 may act as a pro-oxidant during hyperoxia exposure, contributing to increases in oxidative DNA damage and accumulation of lipid hydroperoxides.
Collapse
Affiliation(s)
- Alex C Veith
- Section of Neonatology, Department of Pediatrics.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | | | - Weiwu Jiang
- Section of Neonatology, Department of Pediatrics
| | - Lihua Wang
- Section of Neonatology, Department of Pediatrics
| | - Guodong Zhou
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | | | - Bhagavatula Moorthy
- Section of Neonatology, Department of Pediatrics.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|