1
|
Baer B, Lin J, Schaaf KR, Ware LB, Shaver CM, Bastarache JA. Matrix metalloproteinase-7 is dispensable in a mouse model of sepsis-induced acute lung injury. PLoS One 2025; 20:e0321349. [PMID: 40341670 PMCID: PMC12061409 DOI: 10.1371/journal.pone.0321349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/05/2025] [Indexed: 05/10/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening form of acute lung injury whose pathogenesis is characterized by excessive lung inflammation and alveolar-capillary barrier permeability. Matrix metalloproteinase 7 (MMP7) can regulate leukocyte recruitment and the production of pro-inflammatory cytokines, but whether it plays a role in acute lung injury (ALI) is an unanswered question. We hypothesized that global loss of MMP7 would attenuate sepsis-induced ALI and systemic inflammation. To test this, male and female MMP7 knockout (MMP7KO) mice and wild-type (WT) littermates were exposed to a two-hit model of ALI (sepsis+hyperoxia). Sepsis was induced through intraperitoneal injection of cecal slurry (CS; 1.6mg/g) or 5% dextrose (control) followed by exposure to hyperoxia (HO; FiO2=0.95) or room air (control, FiO2=0.21). At 24-hours post-CS+HO, we measured weight loss, illness severity, and body temperature. The mice were then sacrificed, and samples from the lungs, kidneys, spleen, blood, peritoneal wash, and bronchoalveolar lavage (BAL) fluid were collected for analysis. Bacterial burden was assessed in the peritoneum, lung, and spleen. Lung inflammation was assessed by BAL inflammatory cell recruitment and pro-inflammatory cytokine concentrations as well as lung tissue mRNA expression of pro-inflammatory cytokines. Alveolar-capillary barrier disruption was quantified by BAL total protein, BAL immunoglobulin M, and lung wet-to-dry weight ratios. Histologic evidence of lung injury was evaluated using a histological scoring system. Systemic inflammation was measured through plasma pro-inflammatory cytokines and peritoneal inflammatory cells. Kidney function, inflammation, and injury were assessed through plasma urea nitrogen concentrations, as well as tissue levels of pro-inflammatory cytokines, neutrophil gelatinase-associated lipocalin (NGAL), and kidney injury molecule 1 (KIM-1). Relative mRNA expression of MMP-7, MMP-9, and MMP-2 was also quantified in both lung and kidney tissue through qPCR. At 24-hours post-CS+HO all mice developed ALI. Septic mice also had increased systemic inflammation, kidney inflammation, kidney injury, and kidney dysfunction compared to controls. Loss of MMP7 did not affect markers of inflammation, organ injury, or organ dysfunction. Interestingly, septic male mice exhibited more severe illness, systemic and lung inflammation, lung injury, and lung expression of matrix metalloproteinases, while septic female mice exhibited more kidney inflammation, kidney injury, and kidney expression of matrix metalloproteinases. In conclusion, MMP7 is not essential for the development or resolution of sepsis-induced ALI in this model and likely plays a limited role in the condition.
Collapse
Affiliation(s)
- Brandon Baer
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jason Lin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Kaitlyn R. Schaaf
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Ciara M. Shaver
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Julie A. Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
2
|
Zhou K, Qin Q, Lu J. Pathophysiological mechanisms of ARDS: a narrative review from molecular to organ-level perspectives. Respir Res 2025; 26:54. [PMID: 39948645 PMCID: PMC11827456 DOI: 10.1186/s12931-025-03137-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) remains a life-threatening pulmonary condition with persistently high mortality rates despite significant advancements in supportive care. Its complex pathophysiology involves an intricate interplay of molecular and cellular processes, including cytokine storms, oxidative stress, programmed cell death, and disruption of the alveolar-capillary barrier. These mechanisms drive localized lung injury and contribute to systemic inflammatory response syndrome and multiple organ dysfunction syndrome. Unlike prior reviews that primarily focus on isolated mechanisms, this narrative review synthesizes the key pathophysiological processes of ARDS across molecular, cellular, tissue, and organ levels. MAIN BODY By integrating classical theories with recent research advancements, we provide a comprehensive analysis of how inflammatory mediators, metabolic reprogramming, oxidative stress, and immune dysregulation synergistically drive ARDS onset and progression. Furthermore, we critically evaluate current evidence-based therapeutic strategies, such as lung-protective ventilation and prone positioning, while exploring innovative therapies, including stem cell therapy, gene therapy, and immunotherapy. We emphasize the significance of ARDS subtypes and their inherent heterogeneity in guiding the development of personalized treatment strategies. CONCLUSIONS This narrative review provides fresh perspectives for future research, ultimately enhancing patient outcomes and optimizing management approaches in ARDS.
Collapse
Affiliation(s)
- Kaihuan Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Qianqian Qin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
| |
Collapse
|
3
|
Lavarti R, Cai L, Alvarez‐Diaz T, Medina‐Rodriguez T, Bombin S, Raju RP. Senescence landscape in the liver following sepsis and senolytics as potential therapeutics. Aging Cell 2025; 24:e14354. [PMID: 39444093 PMCID: PMC11709100 DOI: 10.1111/acel.14354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/15/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Senescence, caused by cell-cycle arrest, is a hallmark of aging. Senescence has also been described in embryogenesis, wound healing, and acute injuries. Sepsis is characterized by a dysregulated host response to infection, leading to organ dysfunction and mortality. Most of the pathophysiology of human sepsis is recapitulated in the mouse model of polymicrobial sepsis, developed by cecal ligation and puncture (CLP). In this report, we demonstrate a rapid onset of cellular senescence in the liver of mice subjected to CLP-induced sepsis, characterized by the upregulation of p21, p53, and other senescence markers, including SA-βgal. Using RNAscope, confocal microscopy, and flow cytometry, we further confirm the emergence of p21-expressing senescence phenotype in the liver 24 h after sepsis induction. Senescence was observed in several cell types in the liver, including hepatocytes, endothelial cells, and macrophages. We determined the landscape of senescence phenotype in murine sepsis by single-cell sequencing, which further ascertained that this cell fate is not confined to any particular cell type but displays a heterogeneous distribution. Furthermore, we observed a significant reduction in mortality following sepsis when mice were treated with senolytics, a combination of dasatinib and quercetin, before the CLP surgery. Our experiments unequivocally demonstrated a rapid development of cellular senescence with sepsis and, for the first time, described the senescence landscape in the sepsis liver and the possible role of senescent cells in the worsening outcome following sepsis.
Collapse
Affiliation(s)
- Rupa Lavarti
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Lun Cai
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Tatiana Alvarez‐Diaz
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Thalia Medina‐Rodriguez
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Sergei Bombin
- Georgia Cancer Center, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
4
|
Deshmukh H, Whitsett J, Zacharias W, Way SS, Martinez FD, Mizgerd J, Pryhuber G, Ambalavanan N, Bacharier L, Natarajan A, Tamburro R, Lin S, Randolph A, Nino G, Mejias A, Ramilo O. Impact of Viral Lower Respiratory Tract Infection (LRTI) in Early Childhood (0-2 Years) on Lung Growth and Development and Lifelong Trajectories of Pulmonary Health: A National Institutes of Health (NIH) Workshop Summary. Pediatr Pulmonol 2025; 60:e27357. [PMID: 39565217 PMCID: PMC11740654 DOI: 10.1002/ppul.27357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024]
Abstract
Viral lower respiratory tract infections (LRTI) are ubiquitous in early life. They are disproportionately severe in infants and toddlers (0-2 years), leading to more than 100,000 hospitalizations in the United States per year. The recent relative resilience to severe Coronavirus disease (COVID-19) observed in young children is surprising. These observations, taken together, underscore current knowledge gaps in the pathogenesis of viral lower respiratory tract diseases in young children and respiratory developmental immunology. Further, early-life respiratory viral infections could have a lasting impact on lung development with potential life-long pulmonary sequelae. Modern molecular methods, including high-resolution spatial and single-cell technologies, in concert with longitudinal observational studies beginning in the prenatal period and continuing into early childhood, promise to elucidate developmental pulmonary and immunophenotypes following early-life viral infections and their impact on trajectories of future respiratory health. In November 2019, under the auspices of a multi-disciplinary Workshop convened by the National Heart Lung Blood Institute and the Eunice Kennedy Shriver National Institute of Child Health and Human Development, experts came together to highlight the challenges of respiratory viral infections, particularly in early childhood, and emphasize the knowledge gaps in immune, virological, developmental, and clinical factors that contribute to disease severity and long-term pulmonary morbidity from viral LRTI in children. We hope that the scientific community will view these challenges in clinical care on pulmonary health trajectories and disease burden not as a window of susceptibility but as a window of opportunity.
Collapse
Affiliation(s)
- Hitesh Deshmukh
- Divisions of NeonatologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Pulmonary Biology, and Infectious DiseasesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Medical Scientist Training ProgramUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Jeffrey Whitsett
- Divisions of NeonatologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Pulmonary Biology, and Infectious DiseasesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - William Zacharias
- Pulmonary Biology, and Infectious DiseasesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Medical Scientist Training ProgramUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Sing Sing Way
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Cincinnati Children's Hospital Medical CenterUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Fernando D. Martinez
- Asthma and Airway Disease Research CenterThe University of ArizonaTucsonArizonaUSA
| | - Joseph Mizgerd
- Pulmonary CenterBoston University School of MedicineBostonMassachusettsUSA
| | - Gloria Pryhuber
- Division of Neonatology, Department of Pediatrics, Golisano Children's HospitalUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of PediatricsUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Leonard Bacharier
- Department of PediatricsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | - Robert Tamburro
- Eunice Kennedy Shriver National Institutes of Child Health and Human DevelopmentBethesdaMarylandUSA
| | - Sara Lin
- National Heart, Lung and Blood InstituteBethesdaMarylandUSA
| | - Adrienne Randolph
- Departments of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Departments of Anaesthesia and Harvard Medical SchoolCambridgeMassachusettsUSA
- Pediatrics, Harvard Medical SchoolCambridgeMassachusettsUSA
| | - Gustavo Nino
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National HospitalGeorge Washington UniversityWashingtonD.C.USA
| | - Asuncion Mejias
- Department of Infectious DiseasesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Octavio Ramilo
- Department of Infectious DiseasesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| |
Collapse
Collaborators
N Ambalavanan, Leonard B Bacharier, Gustavo Nino, Bria M Coates, Hitesh Deshmukh, Melody Duvall, Matthew Frieman, James Gern, Sarah Gilpin, Thomas B Kepler, Michele Kong, Richard Locksley, Thomas Mariani, Fernando Martinez, Sharon McGrath-Marrow, Asuncion Mejias, Lisa Miller, Joseph Mizgerd, Wayne Morgan, Peter Mourani, Gloria Pryhuber, Octavio Ramilo, Adrienne Randolph, Fariba Rezaee, Jason Rock, Jeffrey Jeff Whitsett, Sing Sing Way, William Zacharias, Neil Aggarwal, Marishka Brown, Matthew Craig, James Kiley, Sonnie Kim, Marrah Lachowicz-Scroggins, Sara Lin, Valerie Maholmes, Aruna Natarajan, Robert Tamburro,
Collapse
|
5
|
Pereira-Fantini PM, Byars SG, Kamlin COF, Manley BJ, Davis PG, Tingay DG. Plasma Proteome Profiles Associated with Early Development of Lung Injury in Extremely Preterm Infants. Am J Respir Cell Mol Biol 2024; 71:677-687. [PMID: 39051934 DOI: 10.1165/rcmb.2024-0034oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/25/2024] [Indexed: 07/27/2024] Open
Abstract
The biological mediators that initiate lung injury in extremely preterm infants during early postnatal life remain largely unidentified, limiting opportunities for early treatment and diagnosis. In this exploratory study, we used sequential window acquisition of all theoretical mass spectra mass spectrometry to identify bronchopulmonary dysplasia (BPD)-specific changes in protein abundance in plasma samples obtained in the first 72 hours of life from extremely preterm infants and bioinformatic analysis to identify BPD-related biological categories and pathways. Last, binary logistic regression analysis was used to test the BPD predictive potential of a base model alone (gestational age, birth weight, sex) and with the protein biomarker added, with bootstrap resampling used to internally validate protein predictors and adjust for overoptimism. We observed disturbance of key processes, including coagulation, complement activation, development, and extracellular matrix organization, in the first days of life in extremely preterm infants who later received diagnoses of BPD. In the BPD prediction analysis, 49 plasma proteins were identified; when each singularly was combined with birth characteristics the optimism-adjusted C index was 0.65-0.84, suggesting predictive potential for BPD outcomes. Taken together, the results of this study demonstrate that alterations in plasma proteins can be detected from 4 hours of age in extremely preterm infants who later develop BPD and that protein biomarkers, when combined with three birth characteristics, have the potential to predict BPD development within the first 72 hours of life.
Collapse
Affiliation(s)
- Prue M Pereira-Fantini
- Neonatal Research Group and
- Department of Paediatrics and
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - Sean G Byars
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - C Omar F Kamlin
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - Brett J Manley
- Victorian Infant Brain Studies Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - Peter G Davis
- Neonatal Research Group and
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - David G Tingay
- Neonatal Research Group and
- Department of Paediatrics and
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| |
Collapse
|
6
|
Fan W, Gui B, Zhou X, Li L, Chen H. A narrative review on lung injury: mechanisms, biomarkers, and monitoring. Crit Care 2024; 28:352. [PMID: 39482752 PMCID: PMC11526606 DOI: 10.1186/s13054-024-05149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/26/2024] [Indexed: 11/03/2024] Open
Abstract
Lung injury is closely associated with the heterogeneity, severity, mortality, and prognosis of various respiratory diseases. Effective monitoring of lung injury is crucial for the optimal management and improved outcomes of patients with lung diseases. This review describes acute and chronic respiratory diseases characterized by significant lung injury and current clinical tools for assessing lung health. Furthermore, we summarized the mechanisms of lung cell death observed in these diseases and highlighted recently identified biomarkers in the plasma indicative of injury to specific cell types and scaffold structure in the lung. Last, we propose an artificial intelligence-driven lung injury monitoring model to assess disease severity, and predict mortality and prognosis, aiming to achieve precision and personalized medicine.
Collapse
Affiliation(s)
- Wenping Fan
- Department of Respiratory Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China
| | - Biyu Gui
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China
| | - Xiaolei Zhou
- Department of Pulmonary Medicine, Chest Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Li Li
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
| | - Huaiyong Chen
- Department of Respiratory Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China.
- Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
| |
Collapse
|
7
|
Zhou K, Lu J. Progress in cytokine research for ARDS: A comprehensive review. Open Med (Wars) 2024; 19:20241076. [PMID: 39479463 PMCID: PMC11524396 DOI: 10.1515/med-2024-1076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/16/2024] [Accepted: 10/06/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction Acute respiratory distress syndrome (ARDS) is a critical form of acute respiratory failure characterized by diffuse alveolar damage, refractory hypoxemia, and non-cardiogenic pulmonary edema, resulting in high mortality. Dysregulated inflammation, driven by cytokines, is central to ARDS pathogenesis, progression, and prognosis. Objective This review synthesizes current knowledge on the role of cytokines in ARDS and evaluates their potential as therapeutic targets, offering new insights for clinical management. Methods A comprehensive analysis of recent studies was conducted to explore the roles of pro-inflammatory cytokines (e.g., IL-1β, IL-6, IL-8) and anti-inflammatory cytokines (e.g., IL-10, IL-22) in ARDS pathogenesis and to assess current and emerging therapies targeting these cytokines. Results Pro-inflammatory cytokines are crucial in initiating inflammatory responses and lung injury in early ARDS, while anti-inflammatory cytokines help regulate and resolve inflammation. Targeted therapies, such as IL-1 and IL-6 inhibitors, show potential in managing ARDS, particularly in COVID-19, but their clinical efficacy is still debated. Combination therapy strategies may enhance outcomes, but further large-scale, multicenter randomized controlled trials are required to establish their safety and efficacy. Conclusion Understanding cytokine regulation in ARDS could lead to innovative therapeutic approaches. Future research should focus on cytokine roles across ARDS subtypes and stages and develop biomarker-driven, individualized treatments.
Collapse
Affiliation(s)
- Kaihuan Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxuedong Road, Nanning, Guangxi 530007, China
| |
Collapse
|
8
|
Panetti B, Bucci I, Di Ludovico A, Pellegrino GM, Di Filippo P, Di Pillo S, Chiarelli F, Attanasi M, Sferrazza Papa GF. Acute Respiratory Failure in Children: A Clinical Update on Diagnosis. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1232. [PMID: 39457197 PMCID: PMC11506303 DOI: 10.3390/children11101232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
Acute respiratory failure (ARF) is a sudden failure of the respiratory system to ensure adequate gas exchanges. Numerous clinical conditions may cause ARF, including pneumonia, obstructive lung diseases (e.g., asthma), restrictive diseases such as neuromuscular diseases (e.g., spinal muscular atrophy and muscular dystrophy), and albeit rarely, interstitial lung diseases. Children, especially infants, may be more vulnerable to ARF than adults due to anatomical and physiological features of the respiratory system. Assessing respiratory impairment in the pediatric population is particularly challenging as children frequently present difficulties in reporting symptoms and due to compliance and cooperation in diagnostic tests. The evaluation of clinical and anamnestic aspects represents the cornerstone of ARF diagnosis: first level exams (e.g., arterial blood gas analysis) confirm and evaluate the severity of the ARF and second level exams help to uncover the underlying cause. Prompt management is critical, with supplemental oxygen, mechanical ventilation, and the treatment of the underlying problem. The aim of this review is to provide a comprehensive summary of the current state of the art in diagnosing pediatric ARF, with a focus on pathophysiology, novel imaging applications, and new perspectives, such as biomarkers and artificial intelligence.
Collapse
Affiliation(s)
- Beatrice Panetti
- Pediatric Allergy and Pulmonology Unit, Department of Pediatrics, University of Chieti, 66100 Chieti, Italy; (B.P.); (I.B.); (A.D.L.); (P.D.F.); (S.D.P.); (F.C.)
| | - Ilaria Bucci
- Pediatric Allergy and Pulmonology Unit, Department of Pediatrics, University of Chieti, 66100 Chieti, Italy; (B.P.); (I.B.); (A.D.L.); (P.D.F.); (S.D.P.); (F.C.)
| | - Armando Di Ludovico
- Pediatric Allergy and Pulmonology Unit, Department of Pediatrics, University of Chieti, 66100 Chieti, Italy; (B.P.); (I.B.); (A.D.L.); (P.D.F.); (S.D.P.); (F.C.)
| | - Giulia Michela Pellegrino
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, 20144 Milan, Italy; (G.M.P.); (G.F.S.P.)
| | - Paola Di Filippo
- Pediatric Allergy and Pulmonology Unit, Department of Pediatrics, University of Chieti, 66100 Chieti, Italy; (B.P.); (I.B.); (A.D.L.); (P.D.F.); (S.D.P.); (F.C.)
| | - Sabrina Di Pillo
- Pediatric Allergy and Pulmonology Unit, Department of Pediatrics, University of Chieti, 66100 Chieti, Italy; (B.P.); (I.B.); (A.D.L.); (P.D.F.); (S.D.P.); (F.C.)
| | - Francesco Chiarelli
- Pediatric Allergy and Pulmonology Unit, Department of Pediatrics, University of Chieti, 66100 Chieti, Italy; (B.P.); (I.B.); (A.D.L.); (P.D.F.); (S.D.P.); (F.C.)
| | - Marina Attanasi
- Pediatric Allergy and Pulmonology Unit, Department of Pediatrics, University of Chieti, 66100 Chieti, Italy; (B.P.); (I.B.); (A.D.L.); (P.D.F.); (S.D.P.); (F.C.)
| | | |
Collapse
|
9
|
Sallee CJ, Maddux AB, Hippensteel JA, Markovic D, Oshima K, Schwingshackl A, Mourani PM, Schmidt EP, Sapru A. CIRCULATING HEPARAN SULFATE PROFILES IN PEDIATRIC ACUTE RESPIRATORY DISTRESS SYNDROME. Shock 2024; 62:496-504. [PMID: 39331799 PMCID: PMC12080468 DOI: 10.1097/shk.0000000000002421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
ABSTRACT Introduction: Sepsis-induced degradation of endothelial glycocalyx heparan sulfate (HS) contributes to the pulmonary microvascular endothelial injury characteristic of acute respiratory distress syndrome (ARDS) pathogenesis. Our objectives were to (1) examine relationships between plasma indices of HS degradation and protein biomarkers of endothelial injury and (2) identify patient subgroups characterized by distinct profiles of HS degradation in children with ARDS. Methods: We analyzed prospectively collected plasma (2018-2020) from a cohort of invasively mechanically ventilated children (aged >1 month to <18 years) with ARDS. Mass spectrometry characterized and quantified patterns of HS disaccharide sulfation. Protein biomarkers reflective of endothelial injury (e.g., angiopoietin-2, vascular cell adhesion molecule-1, soluble thrombomodulin) were measured with a multiplex immunoassay. Pearson correlation coefficients were used to construct a biomarker correlation network. Centrality metrics detected influential biomarkers (i.e., network hubs). K-means clustering identified unique patient subgroups based on HS disaccharide profiles. Results: We evaluated 36 patients with pediatric ARDS. HS disaccharide sulfation patterns, 6S, NS, and NS2S, positively correlated with all biomarkers of endothelial injury (all P < 0.05) and were classified as network hubs. We identified three patient subgroups, with cluster 3 (n = 5) demonstrating elevated levels of 6S and N-sulfated HS disaccharides. In cluster 3, 60% of children were female and nonpulmonary sepsis accounted for 60% of cases. Relative to cluster 1 (n = 12), cluster 3 was associated with higher oxygen saturation index (P = 0.029) and fewer 28-day ventilator-free days (P = 0.016). Conclusions: Circulating highly sulfated HS fragments may represent emerging mechanistic biomarkers of endothelial injury and disease severity in pediatric ARDS.
Collapse
Affiliation(s)
- Colin J. Sallee
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children’s Hospital, Los Angeles, California
| | - Aline B. Maddux
- Department of Pediatrics, Section of Pediatric Critical Care, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado
| | - Joseph A. Hippensteel
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniela Markovic
- Department of Medicine, Biostatistics Core, University of California Los Angeles, Los Angeles, California
| | - Kaori Oshima
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Andreas Schwingshackl
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children’s Hospital, Los Angeles, California
| | - Peter M. Mourani
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Arkansas for Medical Sciences and Arkansas Children’s Hospital, Little Rock, Arkansas
| | - Eric P. Schmidt
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Anil Sapru
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children’s Hospital, Los Angeles, California
| |
Collapse
|
10
|
Sallee CJ, Hippensteel JA, Miller KR, Oshima K, Pham AT, Richter RP, Belperio J, Sierra YL, Schwingshackl A, Mourani PM, Schmidt EP, Sapru A, Maddux AB. Endothelial Glycocalyx Degradation Patterns in Sepsis-Associated Pediatric Acute Respiratory Distress Syndrome: A Single Center Retrospective Observational Study. J Intensive Care Med 2024; 39:277-287. [PMID: 37670670 PMCID: PMC10845819 DOI: 10.1177/08850666231200162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND Sepsis-associated destruction of the pulmonary microvascular endothelial glycocalyx (EGCX) creates a vulnerable endothelial surface, contributing to the development of acute respiratory distress syndrome (ARDS). Constituents of the EGCX shed into circulation, glycosaminoglycans and proteoglycans, may serve as biomarkers of endothelial dysfunction. We sought to define the patterns of plasma EGCX degradation products in children with sepsis-associated pediatric ARDS (PARDS), and test their association with clinical outcomes. METHODS We retrospectively analyzed a prospective cohort (2018-2020) of children (≥1 month to <18 years of age) receiving invasive mechanical ventilation for acute respiratory failure for ≥72 h. Children with and without sepsis-associated PARDS were selected from the parent cohort and compared. Blood was collected at time of enrollment. Plasma glycosaminoglycan disaccharide class (heparan sulfate, chondroitin sulfate, and hyaluronan) and sulfation subtypes (heparan sulfate and chondroitin sulfate) were quantified using liquid chromatography tandem mass spectrometry. Plasma proteoglycans (syndecan-1) were measured through an immunoassay. RESULTS Among the 39 mechanically ventilated children (29 with and 10 without sepsis-associated PARDS), sepsis-associated PARDS patients demonstrated higher levels of heparan sulfate (median 639 ng/mL [interquartile range, IQR 421-902] vs 311 [IQR 228-461]) and syndecan-1 (median 146 ng/mL [IQR 32-315] vs 8 [IQR 8-50]), both p = 0.01. Heparan sulfate subtype analysis demonstrated greater proportions of N-sulfated disaccharide levels among children with sepsis-associated PARDS (p = 0.01). Increasing N-sulfated disaccharide levels by quartile were associated with severe PARDS (n = 9/29) with the highest quartile including >60% of the severe PARDS patients (test for trend, p = 0.04). Higher total heparan sulfate and N-sulfated disaccharide levels were independently associated with fewer 28-day ventilator-free days in children with sepsis-associated PARDS (all p < 0.05). CONCLUSIONS Children with sepsis-associated PARDS exhibited higher plasma levels of heparan sulfate disaccharides and syndecan-1, suggesting that EGCX degradation biomarkers may provide insights into endothelial dysfunction and PARDS pathobiology.
Collapse
Affiliation(s)
- Colin J. Sallee
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children's Hospital, Los Angeles, CA, USA
| | - Joseph A. Hippensteel
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen R. Miller
- Department of Pediatrics, Section of Pediatric Critical Care, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Kaori Oshima
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Andrew T. Pham
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert P. Richter
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - John Belperio
- Department of Medicine, Division of Pulmonary Critical Care and Sleep Medicine, David Geffen School of Medicine at University of California Los Angeles and Ronald Reagan Medical Center, Los Angeles, CA, USA
| | - Yamila L. Sierra
- Department of Pediatrics, Section of Pediatric Critical Care, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Andreas Schwingshackl
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children's Hospital, Los Angeles, CA, USA
| | - Peter M. Mourani
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, AR, USA
| | - Eric P. Schmidt
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Anil Sapru
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children's Hospital, Los Angeles, CA, USA
| | - Aline B. Maddux
- Department of Pediatrics, Section of Pediatric Critical Care, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
11
|
Yehya N, Zinter MS, Thompson JM, Lim MJ, Hanudel MR, Alkhouli MF, Wong H, Alder MN, McKeone DJ, Halstead ES, Sinha P, Sapru A. Identification of molecular subphenotypes in two cohorts of paediatric ARDS. Thorax 2024; 79:128-134. [PMID: 37813544 PMCID: PMC10850835 DOI: 10.1136/thorax-2023-220130] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Two subphenotypes of acute respiratory distress syndrome (ARDS), hypoinflammatory and hyperinflammatory, have been reported in adults and in a single paediatric cohort. The relevance of these subphenotypes in paediatrics requires further investigation. We aimed to identify subphenotypes in two large observational cohorts of paediatric ARDS and assess their congruence with prior descriptions. METHODS We performed latent class analysis (LCA) separately on two cohorts using biomarkers as inputs. Subphenotypes were compared on clinical characteristics and outcomes. Finally, we assessed overlap with adult cohorts using parsimonious classifiers. FINDINGS In two cohorts from the Children's Hospital of Philadelphia (n=333) and from a multicentre study based at the University of California San Francisco (n=293), LCA identified two subphenotypes defined by differential elevation of biomarkers reflecting inflammation and endotheliopathy. In both cohorts, hyperinflammatory subjects had greater illness severity, more sepsis and higher mortality (41% and 28% in hyperinflammatory vs 11% and 7% in hypoinflammatory). Both cohorts demonstrated overlap with adult subphenotypes when assessed using parsimonious classifiers. INTERPRETATION We identified hypoinflammatory and hyperinflammatory subphenotypes of paediatric ARDS from two separate cohorts with utility for prognostic and potentially predictive, enrichment. Future paediatric ARDS trials should identify and leverage biomarker-defined subphenotypes in their analysis.
Collapse
Affiliation(s)
- Nadir Yehya
- Division of Pediatric Critical Care, Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Matt S Zinter
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jill M Thompson
- Division of Pediatric Critical Care, Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Michelle J Lim
- Department of Pediatrics, UC Davis, Davis, California, USA
| | - Mark R Hanudel
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California, USA
| | - Mustafa F Alkhouli
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Hector Wong
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Matthew N Alder
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Daniel J McKeone
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - E Scott Halstead
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Pratik Sinha
- Division of Clinical and Translational Research, Washington University School of Medicine, St. Louis, MO, USA
- Division of Critical Care, Department of Anesthesia, Washington University, St. Louis, MO, USA
| | - Anil Sapru
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
12
|
Wu M, Wu S, Tan S, Xu Q, Zhang D, Sun J, Yang H, Wang C, Duan T, Xu Y, Wei Z. VitroGel-loaded human MenSCs promote endometrial regeneration and fertility restoration. Front Bioeng Biotechnol 2024; 11:1310149. [PMID: 38260736 PMCID: PMC10800509 DOI: 10.3389/fbioe.2023.1310149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction: Intrauterine adhesions (IUA), also known as Asherman's syndrome, is caused by trauma to the pregnant or non-pregnant uterus, which leads to damaged endometrial basal lining and partial or total occlusion of the uterine chambers, resulting in abnormal menstruation, infertility, or recurrent miscarriage. The essence of this syndrome is endometrial fibrosis. And there is no effective treatment for IUA to stimulate endometrial regeneration currently. Recently, menstrual blood-derived stem cells (MenSCs) have been proved to hold therapeutic promise in various diseases, such as myocardial infarction, stroke, diabetes, and liver cirrhosis. Methods: In this study, we examined the effects of MenSCs on the repair of uterine adhesions in a rat model, and more importantly, promoted such therapeutic effects via a xeno-free VitroGel MMP carrier. Results: This combined treatment reduced the expression of inflammatory factors, increased the expression of anti-inflammatory factors, restricted the area of endometrial fibrosis, diminished uterine adhesions, and partially restored fertility, showing stronger effectiveness than each component alone and almost resembling the sham group. Discussion: Our findings suggest a highly promising strategy for IUA treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tao Duan
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yao Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyun Wei
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
13
|
Williams JG, Jones RL, Yunger TL, Lahni PM, Yehya N, Varisco BM. Comparison of 16 Pediatric Acute Respiratory Distress Syndrome-Associated Plasma Biomarkers With Changing Lung Injury Severity. Pediatr Crit Care Med 2024; 25:e31-e40. [PMID: 37382480 PMCID: PMC10755079 DOI: 10.1097/pcc.0000000000003311] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
OBJECTIVES Pediatric acute respiratory distress syndrome (PARDS) is a source of substantial morbidity and mortality in the PICU, and different plasma biomarkers have identified different PARDS and ARDS subgroups. We have a poor understanding of how these biomarkers change over time and with changing lung injuries. We sought to determine how biomarker levels change over PARDS course, whether they are correlated, and whether they are different in critically ill non-PARDS patients. DESIGN Two-center prospective observational study. SETTING Two quaternary care academic children's hospitals. PATIENTS Subjects under 18 years of age admitted to the PICU who were intubated and met the Second Pediatric Acute Lung Injury Consensus Conference-2 PARDS diagnostic criteria and nonintubated critically ill subjects without apparent lung disease. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Plasma samples were obtained on study days 1, 3, 7, and 14. The levels of 16 biomarkers were measured using a fluorometric bead-based assay. Compared with non-PARDS subjects, on day 1 PARDS subjects had increased concentrations of tumor necrosis factor-alpha, interleukin (IL)-8, interferon-γ, IL17, granzyme B, soluble intercellular adhesion molecule-1 (sICAM1), surfactant protein D, and IL18 but reduced matrix metalloproteinase 9 (MMP-9) concentrations (all p < 0.05). Day 1 biomarker concentrations and PARDS severity were not correlated. Over PARDS course, changes in 11 of the 16 biomarkers positively correlated with changing lung injury with sICAM1 ( R = 0.69, p = 2.2 × 10 -16 ) having the strongest correlation. By Spearman rank correlation of biomarker concentrations in PARDS subjects, we identified two patterns. One had elevations of plasminogen activator inhibitor-1, MMP-9, and myeloperoxidase, and the other had higher inflammatory cytokines. CONCLUSIONS sICAM1 had the strongest positive correlation with worsening lung injury across all study time points suggesting that it is perhaps the most biologically relevant of the 16 analytes. There was no correlation between biomarker concentration on day 1 and day 1 PARDS severity; however, changes in most biomarkers over time positively correlated with changing lung injury. Finally, in day 1 samples, 7 of the 16 biomarkers were not significantly different between PARDS and critically ill non-PARDS subjects. These data highlight the difficulty of using plasma biomarkers to identify organ-specific pathology in critically ill patients.
Collapse
Affiliation(s)
- James G Williams
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Rhonda L Jones
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Toni L Yunger
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Patrick M Lahni
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Nadir Yehya
- Division of Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Brian M Varisco
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
14
|
Liu Y, Huang H, Zhang C, Fan H. N-acetyltransferase 10 promotes the progression of oral squamous cell carcinoma through N4-acetylcytidine RNA acetylation of MMP1 mRNA. Cancer Sci 2023; 114:4202-4215. [PMID: 37705232 PMCID: PMC10637085 DOI: 10.1111/cas.15946] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 09/15/2023] Open
Abstract
The pathogenesis of oral squamous cell carcinoma (OSCC) remains unclear. Therefore, clarifying its pathogenesis and molecular-level development mechanism has become the focus of OSCC research. N-acetyltransferase 10 (NAT10) is a crucial enzyme involved in mRNA acetylation, regulating target gene expression and biological functions of various diseases through mediating N4-acetylcytidine (ac4C) acetylation. However, its role in OSCC progression is not well understood. In this study, we showed that NAT10 was significantly upregulated in OSCC tissues compared to normal oral tissues. Moreover, lentivirus-mediated NAT10 knockdown markedly suppressed cell proliferation, migration, and invasion in two OSCC cell lines (SCC-9 and SCC-15). Interestingly, MMP1 was found to be significantly upregulated in OSCC tissues and was a potential target of NAT10. N-acetyltransferase 10 knockdown significantly reduced both the total and ac4C acetylated levels of MMP1 mRNA and decreased its mRNA stability. Xenograft experiments further confirmed the inhibitory effect of NAT10 knockdown on the tumorigenesis and metastasis ability of OSCC cells and decreased MMP1 expression in vivo. Additionally, NAT10 knockdown impaired the proliferation, migration, and invasion abilities in OSCC cell lines in an MMP1-dependent manner. Our results suggest that NAT10 acts as an oncogene in OSCC, and targeting ac4C acetylation could be a promising therapeutic strategy for OSCC treatment.
Collapse
Affiliation(s)
- Yi Liu
- Department of StomatologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Hui Huang
- Department of StomatologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Cun‐bao Zhang
- Department of Stomatology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineZhejiangChina
| | - Hua‐nan Fan
- Department of StomatologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| |
Collapse
|
15
|
Leimi L, Koski JR, Kilpivaara O, Vettenranta K, Lokki AI, Meri S. Rare variants in complement system genes associate with endothelial damage after pediatric allogeneic hematopoietic stem cell transplantation. Front Immunol 2023; 14:1249958. [PMID: 37771589 PMCID: PMC10525714 DOI: 10.3389/fimmu.2023.1249958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Introduction Complement system has a postulated role in endothelial problems after hematopoietic stem cell transplantation (HSCT). In this retrospective, singlecenter study we studied genetic complement system variants in patients with documented endotheliopathy. In our previous study among pediatric patients with an allogeneic HSCT (2001-2013) at the Helsinki University Children´s Hospital, Finland, we identified a total of 19/122 (15.6%) patients with vascular complications, fulfilling the criteria of capillary leak syndrome (CLS), venoocclusive disease/sinusoidal obstruction syndrome (VOD/SOS) or thrombotic microangiopathy (TMA). Methods We performed whole exome sequencing (WES) on 109 patients having an adequate pre-transplantation DNA for the analysis to define possible variations and mutations potentially predisposing to functional abnormalities of the complement system. In our data analysis, we focused on 41 genes coding for complement components. Results 50 patients (45.9%) had one or several, nonsynonymous, rare germline variants in complement genes. 21/66 (31.8%) of the variants were in the terminal pathway. Patients with endotheliopathy had variants in different complement genes: in the terminal pathway (C6 and C9), lectin pathway (MASP1) and receptor ITGAM (CD11b, part of CR3). Four had the same rare missense variant (rs183125896; Thr279Ala) in the C9 gene. Two of these patients were diagnosed with endotheliopathy and one with capillary leak syndrome-like problems. The C9 variant Thr279Ala has no previously known disease associations and is classified by the ACMG guidelines as a variant of uncertain significance (VUS). We conducted a gene burden test with gnomAD Finnish (fin) as the reference population. Complement gene variants seen in our patient population were investigated and Total Frequency Testing (TFT) was used for execution of burden tests. The gene variants seen in our patients with endotheliopathy were all significantly (FDR < 0.05) enriched compared to gnomAD. Overall, 14/25 genes coding for components of the complement system had an increased burden of missense variants among the patients when compared to the gnomAD Finnish population (N=10 816). Discussion Injury to the vascular endothelium is relatively common after HSCT with different phenotypic appearances suggesting yet unidentified underlying mechanisms. Variants in complement components may be related to endotheliopathy and poor prognosis in these patients.
Collapse
Affiliation(s)
- Lilli Leimi
- Pediatric Research Center, Children’s Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Jessica R. Koski
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Outi Kilpivaara
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Kim Vettenranta
- Pediatric Research Center, Children’s Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - A. Inkeri Lokki
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Seppo Meri
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Makieieva N, Koval V, Kondratiuk T, Gorbach T. 1. CHILD`S HEALTH 2023; 18:194-200. [DOI: 10.22141/2224-0551.18.3.2023.1585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background. Endothelial dysfunction (ED) is common in acute leukemia patients. The study of ED can provide more information about pathological processes in lungs of children with acute lymphoblastic leukemia (ALL). The purpose of the study is to assess the levels of vascular endothelial growth factor A (VEGF-A) and its prognostic value for pulmonary complications in children with ALL. Materials and methods. The control group consisted of 15 healthy children. The level of VEGF-A in serum was assessed by enzyme-linked immunosorbent assay. Results. Pulmonary complications were common in the examined children with ALL, among them: аcute bronchitis (23), recurrent episodes of acute bronchitis (5), pneumonia (18), wheezing (9), bronchial asthma (3), interstitial pneumonia (1), pleurisy (1), pneumothorax (3), lung fibrosis (2), respiratory failure (6). The frequency of pulmonary complications was 82.5 % during chemotherapy protocols and 20.0 % in ALL survivors after a complete course of chemotherapy. Statistically significant increase in VEGF-A level in groups 1 (180.41 (158.16; 200.00) pg/ml) and 2 (165.61 (131.65; 198.45) pg/ml) compared to controls (130.65 (129.45; 132.15) pg/ml) has been detected (p1-C = 0.000011; p2-C = 0.007009). There were no significant differences in VEGF-А levels between children from experimental groups (p1–2 = 0.338394). According to receiver operator characteristic (ROC) analysis, the level of VEGF-A > 198.34 pg/ml after the complete course of chemotherapy can predict the presence of pulmonary complication in ALL survivors (area under the ROC curve 0.965; sensitivity 100.00 %; specificity 89.47 %). Conclusions. Children with ALL have significant ED. The level of serum VEGF-A can be predictive for pulmonary complications in ALL survivors.
Collapse
|
17
|
Jones TW, Almuntashiri S, Chase A, Alhumaid A, Somanath PR, Sikora A, Zhang D. Plasma matrix metalloproteinase-3 predicts mortality in acute respiratory distress syndrome: a biomarker analysis of a randomized controlled trial. Respir Res 2023; 24:166. [PMID: 37349704 PMCID: PMC10286483 DOI: 10.1186/s12931-023-02476-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Matrix metalloproteinase-3 (MMP-3) is a proteolytic enzyme involved in acute respiratory distress syndrome (ARDS) pathophysiology that may serve as a lung-specific biomarker in ARDS. METHODS This study was a secondary biomarker analysis of a subset of Albuterol for the Treatment of Acute Lung Injury (ALTA) trial patients to determine the prognostic value of MMP-3. Plasma sample MMP-3 was measured by enzyme-linked immunosorbent assay. The primary outcome was the area under the receiver operating characteristic (AUROC) of MMP-3 at day 3 for the prediction of 90-day mortality. RESULTS A total of 100 unique patient samples were evaluated and the AUROC analysis of day three MMP-3 showed an AUROC of 0.77 for the prediction of 90-day mortality (95% confidence interval: 0.67-0.87), corresponding to a sensitivity of 92% and specificity of 63% and an optimal cutoff value of 18.4 ng/mL. Patients in the high MMP-3 group (≥ 18.4 ng/mL) showed higher mortality compared to the non-elevated MMP-3 group (< 18.4 ng/mL) (47% vs. 4%, p < 0.001). A positive difference in day zero and day three MMP-3 concentration was predictive of mortality with an AUROC of 0.74 correlating to 73% sensitivity, 81% specificity, and an optimal cutoff value of + 9.5 ng/mL. CONCLUSIONS Day three MMP-3 concentration and difference in day zero and three MMP-3 concentrations demonstrated acceptable AUROCs for predicting 90-day mortality with a cut-point of 18.4 ng/mL and + 9.5 ng/mL, respectively. These results suggest a prognostic role of MMP-3 in ARDS.
Collapse
Affiliation(s)
- Timothy W. Jones
- Department of Pharmacy, Augusta University Medical Center, 1120 15th St., Augusta, GA 30912 USA
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Sultan Almuntashiri
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Aaron Chase
- Department of Pharmacy, Augusta University Medical Center, 1120 15th St., Augusta, GA 30912 USA
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Abdullah Alhumaid
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Payaningal R. Somanath
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Andrea Sikora
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Duo Zhang
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| |
Collapse
|
18
|
Feng F, Wang LJ, Li JC, Chen TT, Liu L. Role of heparanase in ARDS through autophagy and exosome pathway (review). Front Pharmacol 2023; 14:1200782. [PMID: 37361227 PMCID: PMC10285077 DOI: 10.3389/fphar.2023.1200782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is the most common respiratory disease in ICU. Although there are many treatment and support methods, the mortality rate is still high. The main pathological feature of ARDS is the damage of pulmonary microvascular endothelium and alveolar epithelium caused by inflammatory reaction, which may lead to coagulation system disorder and pulmonary fibrosis. Heparanase (HPA) plays an significant role in inflammation, coagulation, fibrosis. It is reported that HPA degrades a large amount of HS in ARDS, leading to the damage of endothelial glycocalyx and inflammatory factors are released in large quantities. HPA can aggrandize the release of exosomes through syndecan-syntenin-Alix pathway, leading to a series of pathological reactions; at the same time, HPA can cause abnormal expression of autophagy. Therefore, we speculate that HPA promotes the occurrence and development of ARDS through exosomes and autophagy, which leads to a large amount of release of inflammatory factors, coagulation disorder and pulmonary fibrosis. This article mainly describes the mechanism of HPA on ARDS.
Collapse
Affiliation(s)
- Fei Feng
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Lin-Jun Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Jian-Chun Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Ting-Ting Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Liping Liu
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
19
|
Pathobiology, Severity, and Risk Stratification of Pediatric Acute Respiratory Distress Syndrome: From the Second Pediatric Acute Lung Injury Consensus Conference. Pediatr Crit Care Med 2023; 24:S12-S27. [PMID: 36661433 DOI: 10.1097/pcc.0000000000003156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVES To review the literature for studies published in children on the pathobiology, severity, and risk stratification of pediatric acute respiratory distress syndrome (PARDS) with the intent of guiding current medical practice and identifying important areas for future research related to severity and risk stratification. DATA SOURCES Electronic searches of PubMed and Embase were conducted from 2013 to March 2022 by using a combination of medical subject heading terms and text words to capture the pathobiology, severity, and comorbidities of PARDS. STUDY SELECTION We included studies of critically ill patients with PARDS that related to the severity and risk stratification of PARDS using characteristics other than the oxygenation defect. Studies using animal models, adult only, and studies with 10 or fewer children were excluded from our review. DATA EXTRACTION Title/abstract review, full-text review, and data extraction using a standardized data collection form. DATA SYNTHESIS The Grading of Recommendations Assessment, Development, and Evaluation approach was used to identify and summarize relevant evidence and develop recommendations for clinical practice. There were 192 studies identified for full-text extraction to address the relevant Patient/Intervention/Comparator/Outcome questions. One clinical recommendation was generated related to the use of dead space fraction for risk stratification. In addition, six research statements were generated about the impact of age on acute respiratory distress syndrome pathobiology and outcomes, addressing PARDS heterogeneity using biomarkers to identify subphenotypes and endotypes, and use of standardized ventilator, physiologic, and nonpulmonary organ failure measurements for future research. CONCLUSIONS Based on an extensive literature review, we propose clinical management and research recommendations related to characterization and risk stratification of PARDS severity.
Collapse
|
20
|
Yao RQ, Shen Z, Ma QM, Ling P, Wei CR, Zheng LY, Duan Y, Li W, Zhu F, Sun Y, Wu GS. Combination of transcriptional biomarkers and clinical parameters for early prediction of sepsis indued acute respiratory distress syndrome. Front Immunol 2023; 13:1084568. [PMID: 36685531 PMCID: PMC9846102 DOI: 10.3389/fimmu.2022.1084568] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023] Open
Abstract
Objective As a common yet intractable complication of severe sepsis, acute respiratory distress syndrome (ARDS) is closely associated with poor clinical outcomes and elevated medical expenses. The aim of the current study is to generate a model combining transcriptional biomarkers and clinical parameters to alarm the development of ARDS in septic patients. Methods Gene expression profile (GSE66890) was downloaded from the Gene Expression Omnibus database and clinical data were extracted. Differentially expressed genes (DEGs) from whole blood leukocytes were identified between patients with sepsis alone and septic patients who develop ARDS. ARDS prediction model was constructed using backward stepwise regression and Akaike Information Criterion (AIC). Meanwhile, a nomogram based on this model was established, with subsequent internal validation. Results A total of 57 severe septic patients were enrolled in this study, and 28 (49.1%) developed ARDS. Based on the differential expression analysis, six DEGs (BPI, OLFM4, LCN2, CD24, MMP8 and MME) were screened. According to the outcome prediction model, six valuable risk factors (direct lung injury, shock, tumor, BPI, MME and MMP8) were incorporated into a nomogram, which was used to predict the onset of ARDS in septic patients. The calibration curves of the nomogram showed good consistency between the probabilities and observed values. The decision curve analysis also revealed the potential clinical usefulness of the nomogram. The area under the receiver operating characteristic (AUROC) for the prediction of ARDS occurrence in septic patients by the nomogram was 0.86 (95% CI = 0.767-0.952). A sensitivity analysis showed that the AUROC for the prediction of ARDS development in septic patients without direct lung injury was 0.967 (95% CI = 0.896-1.0). Conclusions The nomogram based on transcriptional biomarkers and clinical parameters showed a good performance for the prediction of ARDS occurrence in septic patients.
Collapse
Affiliation(s)
- Ren-Qi Yao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China,Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zong Shen
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Qi-Min Ma
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ping Ling
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chen-Ru Wei
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yu Duan
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Wei Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Feng Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yu Sun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Guo-Sheng Wu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China,*Correspondence: Guo-Sheng Wu,
| |
Collapse
|
21
|
Ripple MJ, Mohammad AF, Stephenson ST, Fitzpatrick AM, Grunwell JR. Expression Patterns of Airway Fluid Cytokines From Intubated Children With Pediatric Acute Respiratory Distress Syndrome. Crit Care Explor 2022; 4:e0819. [PMID: 36567781 PMCID: PMC9760621 DOI: 10.1097/cce.0000000000000819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pediatric acute respiratory distress syndrome (PARDS) is a heterogeneous illness affecting 6% of mechanically ventilated children and with an overall mortality of 17%. Studies in PARDS have mainly focused on plasma biomarkers which may not reflect airway biomarkers. We lack adequate understanding of the inflammatory mediators and underlying immune responses in the airways of PARDS patients. Our objective was to compare the levels of cytokines in the airway fluid of intubated children with severe versus nonsevere acute respiratory distress syndrome. DESIGN Prospective observational cohort study. SETTING Single 36-bed quaternary care academic safety-net hospital PICU. PATIENTS Children intubated for acute respiratory failure between January 2018 and November 2021 stratified by Pediatric Acute Lung Injury Consensus Conference-1 criteria for PARDS. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We measured levels of 23 cytokines, chemokines, and protein biomarkers in the tracheal aspirate from 82 intubated children, between 14 days and 17 years old, at risk for or with PARDS. Levels of interleukin-4, -5, -7, -8, -12(p-70), -17a, -21, and fractalkine were higher in patients with severe versus nonsevere PARDS. There were no associations between airway and plasma cytokines. CONCLUSIONS Proinflammatory cytokines are elevated in the airway fluid from intubated children with severe PARDS and reflect diverse patterns of airway inflammation.
Collapse
Affiliation(s)
- Michael J Ripple
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Division of Critical Care Medicine, Children's Healthcare of Atlanta, Atlanta, GA
| | - Ahmad F Mohammad
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Susan T Stephenson
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Anne M Fitzpatrick
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Division of Pulmonary Medicine, Children's Healthcare of Atlanta, Atlanta, GA
| | - Jocelyn R Grunwell
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Division of Critical Care Medicine, Children's Healthcare of Atlanta, Atlanta, GA
| |
Collapse
|
22
|
Whitney JE, Lee IH, Lee JW, Kong SW. Evolution of multiple omics approaches to define pathophysiology of pediatric acute respiratory distress syndrome. eLife 2022; 11:77405. [PMID: 35913450 PMCID: PMC9342956 DOI: 10.7554/elife.77405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022] Open
Abstract
Pediatric acute respiratory distress syndrome (PARDS), though both common and deadly in critically ill children, lacks targeted therapies. The development of effective pharmacotherapies has been limited, in part, by lack of clarity about the pathobiology of pediatric ARDS. Epithelial lung injury, vascular endothelial activation, and systemic immune activation are putative drivers of this complex disease process. Prior studies have used either hypothesis-driven (e.g., candidate genes and proteins, in vitro investigations) or unbiased (e.g., genome-wide association, transcriptomic, metabolomic) approaches to predict clinical outcomes and to define subphenotypes. Advances in multiple omics technologies, including genomics, transcriptomics, proteomics, and metabolomics, have permitted more comprehensive investigation of PARDS pathobiology. However, omics studies have been limited in children compared to adults, and analyses across multiple tissue types are lacking. Here, we synthesized existing literature on the molecular mechanism of PARDS, summarized our interrogation of publicly available genomic databases to determine the association of candidate genes with PARDS phenotypes across multiple tissues and cell types, and integrated recent studies that used single-cell RNA sequencing (scRNA-seq). We conclude that novel profiling methods such as scRNA-seq, which permits more comprehensive, unbiased evaluation of pathophysiological mechanisms across tissue and cell types, should be employed to investigate the molecular mechanisms of PRDS toward the goal of identifying targeted therapies.
Collapse
Affiliation(s)
- Jane E Whitney
- Medical Critical Care, Pediatrics, Boston Children's Hospital, Boston, United States.,Department of Pediatrics, Harvard Medical School, Boston, United States
| | - In-Hee Lee
- Computational Health and Informatics Program, Boston Children's Hospital, Boston, United States
| | - Ji-Won Lee
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Sek Won Kong
- Department of Pediatrics, Harvard Medical School, Boston, United States.,Computational Health and Informatics Program, Boston Children's Hospital, Boston, United States
| |
Collapse
|
23
|
Liu D, Wen L, Wang Z, Hai Y, Yang D, Zhang Y, Bai M, Song B, Wang Y. The Mechanism of Lung and Intestinal Injury in Acute Pancreatitis: A Review. Front Med (Lausanne) 2022; 9:904078. [PMID: 35872761 PMCID: PMC9301017 DOI: 10.3389/fmed.2022.904078] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP), as a common cause of clinical acute abdomen, often leads to multi-organ damage. In the process of severe AP, the lungs and intestines are the most easily affected organs aside the pancreas. These organ damages occur in succession. Notably, lung and intestinal injuries are closely linked. Damage to ML, which transports immune cells, intestinal fluid, chyle, and toxic components (including toxins, trypsin, and activated cytokines to the systemic circulation in AP) may be connected to AP. This process can lead to the pathological changes of hyperosmotic edema of the lung, an increase in alveolar fluid level, destruction of the intestinal mucosal structure, and impairment of intestinal mucosal permeability. The underlying mechanisms of the correlation between lung and intestinal injuries are inflammatory response, oxidative stress, and endocrine hormone secretion disorders. The main signaling pathways of lung and intestinal injuries are TNF-α, HMGB1-mediated inflammation amplification effect of NF-κB signal pathway, Nrf2/ARE oxidative stress response signaling pathway, and IL-6-mediated JAK2/STAT3 signaling pathway. These pathways exert anti-inflammatory response and anti-oxidative stress, inhibit cell proliferation, and promote apoptosis. The interaction is consistent with the traditional Chinese medicine theory of the lung being connected with the large intestine (fei yu da chang xiang biao li in Chinese). This review sought to explore intersecting mechanisms of lung and intestinal injuries in AP to develop new treatment strategies.
Collapse
Affiliation(s)
- Dongling Liu
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Linlin Wen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- County People’s Hospital, Pingliang, China
| | - Zhandong Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yang Hai
- Gansu University of Chinese Medicine/Scientific Research and Experimental Center, Lanzhou, China
| | - Dan Yang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yanying Zhang
- Gansu University of Chinese Medicine/Scientific Research and Experimental Center, Lanzhou, China
- Gansu Provincial Engineering Laboratory for Research and Promotion of Quality Standardization of Authentic Medicinal Materials in Gansu Province/Provincial Key Laboratory of Pharmaceutical Chemistry and Quality Research in Colleges and Universities in Gansu Province/Gansu Provincial Laboratory Animal Industry Technology Center, Lanzhou, China
| | - Min Bai
- Gansu Provincial Engineering Laboratory for Research and Promotion of Quality Standardization of Authentic Medicinal Materials in Gansu Province/Provincial Key Laboratory of Pharmaceutical Chemistry and Quality Research in Colleges and Universities in Gansu Province/Gansu Provincial Laboratory Animal Industry Technology Center, Lanzhou, China
| | - Bing Song
- Gansu University of Chinese Medicine/Scientific Research and Experimental Center, Lanzhou, China
- Gansu Provincial Engineering Laboratory for Research and Promotion of Quality Standardization of Authentic Medicinal Materials in Gansu Province/Provincial Key Laboratory of Pharmaceutical Chemistry and Quality Research in Colleges and Universities in Gansu Province/Gansu Provincial Laboratory Animal Industry Technology Center, Lanzhou, China
| | - Yongfeng Wang
- Gansu Provincial Engineering Laboratory for Research and Promotion of Quality Standardization of Authentic Medicinal Materials in Gansu Province/Provincial Key Laboratory of Pharmaceutical Chemistry and Quality Research in Colleges and Universities in Gansu Province/Gansu Provincial Laboratory Animal Industry Technology Center, Lanzhou, China
| |
Collapse
|
24
|
Lim MJ, Zinter MS, Chen L, Wong KMY, Bhalla A, Gala K, Guglielmo M, Alkhouli M, Huard LL, Hanudel MR, Vangala S, Schwingshackl A, Matthay M, Sapru A. Beyond the Alveolar Epithelium: Plasma Soluble Receptor for Advanced Glycation End Products Is Associated With Oxygenation Impairment, Mortality, and Extrapulmonary Organ Failure in Children With Acute Respiratory Distress Syndrome. Crit Care Med 2022; 50:837-847. [PMID: 34678846 PMCID: PMC9035468 DOI: 10.1097/ccm.0000000000005373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Soluble receptor for advanced glycation end products is a known plasma marker of alveolar epithelial injury. However, RAGE is also expressed on cell types beyond the lung, and its activation leads to up-regulation of pro-inflammatory mediators. We sought to examine the relationship between plasma soluble receptor for advanced glycation end products and primary pulmonary dysfunction, extrapulmonary organ dysfunction, and mortality in pediatric acute respiratory distress syndrome patients at two early time points following acute respiratory distress syndrome diagnosis and compare these results to plasma surfactant protein-D, a marker of pure alveolar epithelial injury. DESIGN Prospective observational study. SETTING Five academic PICUs. PATIENTS Two hundred fifty-eight pediatric patients 30 days to 18 years old meeting Berlin Criteria for acute respiratory distress syndrome. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Plasma was collected for soluble receptor for advanced glycation end products and surfactant protein-D measurements within 24 hours (day 1) and 48 to 72 hours (day 3) after acute respiratory distress syndrome diagnosis. Similar to surfactant protein-D, plasma soluble receptor for advanced glycation end products was associated with a higher oxygenation index (p < 0.01) and worse lung injury score (p < 0.001) at the time of acute respiratory distress syndrome diagnosis. However, unlike surfactant protein-D, plasma soluble receptor for advanced glycation end products was associated with worse extrapulmonary Pediatric Logistic Organ Dysfunction score during ICU stay (day 3; p < 0.01) and positively correlated with plasma levels of interleukin-6 (p < 0.01), tumor necrosis factor-α (p < 0.01), and angiopoietin-2 (p < 0.01). Among children with indirect lung injury, plasma soluble receptor for advanced glycation end products was associated with mortality independent of age, sex, race, cancer/bone marrow transplant, and Pediatric Risk of Mortality score (day 3; odds ratio, 3.14; 95% CI, 1.46-6.75; p < 0.01). CONCLUSIONS Unlike surfactant protein-D, which is primarily localized to the alveolar epithelium plasma soluble receptor for advanced glycation end products is systemically expressed and correlates with markers of inflammation, extrapulmonary multiple organ dysfunction, and death in pediatric acute respiratory distress syndrome with indirect lung injury. This suggests that unlike surfactant protein-D, soluble receptor for advanced glycation end products is a multifaceted marker of alveolar injury and increased inflammation and that receptor for advanced glycation end products activation may contribute to the pathogenesis of multiple organ failure among children with indirect acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Michelle J. Lim
- UC Davis School of Medicine, UC Davis Children’s Hospital, Department of Pediatrics, Division of Critical Care, Sacramento, CA, USA
| | - Matt S. Zinter
- UCSF School of Medicine, Benioff Children’s Hospital, Department of Pediatrics, Division of Critical Care, San Francisco, CA, USA
| | - Lucia Chen
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Kayley Man Yee Wong
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Anoopindar Bhalla
- USC Keck School of Medicine, Children’s Hospital Los Angeles, Department of Anesthesiology and Critical Care Medicine, Los Angeles, CA, USA
| | - Kinisha Gala
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Mona Guglielmo
- Loma Linda University School of Medicine, Loma Linda University Children’s Hospital, Department of Pediatrics, Division of Critical Care, Loma Linda, CA, USA
| | - Mustafa Alkhouli
- UCSF School of Medicine, Benioff Children’s Hospital, Department of Pediatrics, Division of Critical Care, San Francisco, CA, USA
| | - Leanna L. Huard
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Mark R. Hanudel
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Nephrology, Los Angeles, CA, USA
| | - Sitaram Vangala
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Andreas Schwingshackl
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Michael Matthay
- Departments of Medicine and Anesthesia and the Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Anil Sapru
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| |
Collapse
|
25
|
Mohammed Y, Goodlett DR, Cheng MP, Vinh DC, Lee TC, Mcgeer A, Sweet D, Tran K, Lee T, Murthy S, Boyd JH, Singer J, Walley KR, Patrick DM, Quan C, Ismail S, Amar L, Pal A, Bassawon R, Fesdekjian L, Gou K, Lamontagne F, Marshall J, Haljan G, Fowler R, Winston BW, Russell JA, ARBs CORONA I. Longitudinal Plasma Proteomics Analysis Reveals Novel Candidate Biomarkers in Acute COVID-19. J Proteome Res 2022; 21:975-992. [PMID: 35143212 PMCID: PMC8864781 DOI: 10.1021/acs.jproteome.1c00863] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Indexed: 12/15/2022]
Abstract
The host response to COVID-19 pathophysiology over the first few days of infection remains largely unclear, especially the mechanisms in the blood compartment. We report on a longitudinal proteomic analysis of acute-phase COVID-19 patients, for which we used blood plasma, multiple reaction monitoring with internal standards, and data-independent acquisition. We measured samples on admission for 49 patients, of which 21 had additional samples on days 2, 4, 7, and 14 after admission. We also measured 30 externally obtained samples from healthy individuals for comparison at baseline. The 31 proteins differentiated in abundance between acute COVID-19 patients and healthy controls belonged to acute inflammatory response, complement activation, regulation of inflammatory response, and regulation of protein activation cascade. The longitudinal analysis showed distinct profiles revealing increased levels of multiple lipid-associated functions, a rapid decrease followed by recovery for complement activation, humoral immune response, and acute inflammatory response-related proteins, and level fluctuation in the regulation of smooth muscle cell proliferation, secretory mechanisms, and platelet degranulation. Three proteins were differentiated between survivors and nonsurvivors. Finally, increased levels of fructose-bisphosphate aldolase B were determined in patients with exposure to angiotensin receptor blockers versus decreased levels in those exposed to angiotensin-converting enzyme inhibitors. Data are available via ProteomeXchange PXD029437.
Collapse
Affiliation(s)
- Yassene Mohammed
- Genome BC Proteomics Centre, University
of Victoria, Victoria V8Z 5N3, British Columbia,
Canada
- Center for Proteomics and Metabolomics,
Leiden University Medical Center, Leiden 2333 ZA,
Netherlands
| | - David R. Goodlett
- Genome BC Proteomics Centre, University
of Victoria, Victoria V8Z 5N3, British Columbia,
Canada
- Department of Biochemistry and Microbiology,
University of Victoria, Victoria V8W 2Y2, British Columbia,
Canada
- International Centre for Cancer Vaccine Science,
University of Gdansk, Gdansk 80-822, European Union,
Poland
| | - Matthew P. Cheng
- Division of Infectious Diseases (Department of
Medicine), Division of Medical Microbiology (Department of Pathology and Laboratory
Medicine), McGill University Health Centre, Montreal H4A 3J1,
Quebec, Canada
| | - Donald C. Vinh
- Division of Infectious Diseases (Department of
Medicine), Division of Medical Microbiology (Department of Pathology and Laboratory
Medicine), McGill University Health Centre, Montreal H4A 3J1,
Quebec, Canada
| | - Todd C. Lee
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
| | - Allison Mcgeer
- Mt. Sinai Hospital and University of
Toronto, University Avenue, Toronto M5G 1X5, Ontario,
Canada
| | - David Sweet
- Division of Critical Care Medicine, Department of
Emergency Medicine, Vancouver General Hospital and University of British
Columbia, Vancouver V5Z 1M9, British Columbia,
Canada
| | - Karen Tran
- Division of General Internal Medicine,
Vancouver General Hospital and University of British
Columbia, Vancouver V5Z 1M9, British Columbia,
Canada
| | - Terry Lee
- Centre for Health Evaluation and Outcome Science
(CHEOS), St. Paul’s Hospital, University of British
Columbia, 1081 Burrard Street, Vancouver V6Z 1Y6, British Columbia,
Canada
| | - Srinivas Murthy
- BC Children’s Hospital,
University of British Columbia, Vancouver V6H 3N1, British Columbia,
Canada
| | - John H. Boyd
- Centre for Heart Lung Innovation, St.
Paul’s Hospital, University of British Columbia, 1081 Burrard
Street, Vancouver V6Z 1Y6, British Columbia, Canada
- Division of Critical Care Medicine, St.
Paul’s Hospital, University of British Columbia, 1081 Burrard
Street, Vancouver V6Z 1Y6, British Columbia, Canada
| | - Joel Singer
- Centre for Health Evaluation and Outcome Science
(CHEOS), St. Paul’s Hospital, University of British
Columbia, 1081 Burrard Street, Vancouver V6Z 1Y6, British Columbia,
Canada
| | - Keith R. Walley
- Centre for Heart Lung Innovation, St.
Paul’s Hospital, University of British Columbia, 1081 Burrard
Street, Vancouver V6Z 1Y6, British Columbia, Canada
- Division of Critical Care Medicine, St.
Paul’s Hospital, University of British Columbia, 1081 Burrard
Street, Vancouver V6Z 1Y6, British Columbia, Canada
| | - David M. Patrick
- British Columbia Centre for Disease
Control (BCCDC) and University of British Columbia, Vancouver V5Z 4R4,
British Columbia, Canada
| | - Curtis Quan
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
| | - Sara Ismail
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
| | - Laetitia Amar
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
| | - Aditya Pal
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
| | - Rayhaan Bassawon
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
| | - Lara Fesdekjian
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
| | - Karine Gou
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
| | | | - John Marshall
- Department of Surgery, St.
Michael’s Hospital, Toronto M5B 1W8, Ontario,
Canada
| | - Greg Haljan
- Division of Critical Care, Surrey
Memorial Hospital and University of British Columbia, Surrey V3V 1Z2,
British Columbia, Canada
| | - Robert Fowler
- Sunnybrook Health Sciences
Centre, Toronto M4N 3M5, Ontario, Canada
| | - Brent W. Winston
- Departments of Critical Care Medicine, Medicine and
Biochemistry and Molecular Biology, University of Calgary,
Calgary T2N 4N1, Alberta, Canada
| | - James A. Russell
- Centre for Heart Lung Innovation, St.
Paul’s Hospital, University of British Columbia, 1081 Burrard
Street, Vancouver V6Z 1Y6, British Columbia, Canada
- Division of Critical Care Medicine, St.
Paul’s Hospital, University of British Columbia, 1081 Burrard
Street, Vancouver V6Z 1Y6, British Columbia, Canada
| | - ARBs CORONA I
- Genome BC Proteomics Centre, University
of Victoria, Victoria V8Z 5N3, British Columbia,
Canada
- Center for Proteomics and Metabolomics,
Leiden University Medical Center, Leiden 2333 ZA,
Netherlands
- Department of Biochemistry and Microbiology,
University of Victoria, Victoria V8W 2Y2, British Columbia,
Canada
- International Centre for Cancer Vaccine Science,
University of Gdansk, Gdansk 80-822, European Union,
Poland
- Department of Medicine, McGill
University, Montreal H4A 3J1, Quebec, Canada
- Mt. Sinai Hospital and University of
Toronto, University Avenue, Toronto M5G 1X5, Ontario,
Canada
- Division of Critical Care Medicine, Department of
Emergency Medicine, Vancouver General Hospital and University of British
Columbia, Vancouver V5Z 1M9, British Columbia,
Canada
- Division of General Internal Medicine,
Vancouver General Hospital and University of British
Columbia, Vancouver V5Z 1M9, British Columbia,
Canada
- Centre for Health Evaluation and Outcome Science
(CHEOS), St. Paul’s Hospital, University of British
Columbia, 1081 Burrard Street, Vancouver V6Z 1Y6, British Columbia,
Canada
- BC Children’s Hospital,
University of British Columbia, Vancouver V6H 3N1, British Columbia,
Canada
- Centre for Heart Lung Innovation, St.
Paul’s Hospital, University of British Columbia, 1081 Burrard
Street, Vancouver V6Z 1Y6, British Columbia, Canada
- Division of Critical Care Medicine, St.
Paul’s Hospital, University of British Columbia, 1081 Burrard
Street, Vancouver V6Z 1Y6, British Columbia, Canada
- British Columbia Centre for Disease
Control (BCCDC) and University of British Columbia, Vancouver V5Z 4R4,
British Columbia, Canada
- University of Sherbrooke,
Sherbrooke J1K 2R1, Quebec, Canada
- Department of Surgery, St.
Michael’s Hospital, Toronto M5B 1W8, Ontario,
Canada
- Division of Critical Care, Surrey
Memorial Hospital and University of British Columbia, Surrey V3V 1Z2,
British Columbia, Canada
- Sunnybrook Health Sciences
Centre, Toronto M4N 3M5, Ontario, Canada
- Departments of Critical Care Medicine, Medicine and
Biochemistry and Molecular Biology, University of Calgary,
Calgary T2N 4N1, Alberta, Canada
- Division of Infectious Diseases (Department of
Medicine), Division of Medical Microbiology (Department of Pathology and Laboratory
Medicine), McGill University Health Centre, Montreal H4A 3J1,
Quebec, Canada
| |
Collapse
|
26
|
Conrad C, Yildiz D, Cleary SJ, Margraf A, Cook L, Schlomann U, Panaretou B, Bowser JL, Karmouty-Quintana H, Li J, Berg NK, Martin SC, Aljohmani A, Moussavi-Harami SF, Wang KM, Tian JJ, Magnen M, Valet C, Qiu L, Singer JP, Eltzschig HK, CAPSys Study Group, Bertrams W, Herold S, Suttorp N, Schmeck B, Ball ZT, Zarbock A, Looney MR, Bartsch JW. ADAM8 signaling drives neutrophil migration and ARDS severity. JCI Insight 2022; 7:e149870. [PMID: 35132956 PMCID: PMC8855804 DOI: 10.1172/jci.insight.149870] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 12/21/2021] [Indexed: 01/27/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) results in catastrophic lung failure and has an urgent, unmet need for improved early recognition and therapeutic development. Neutrophil influx is a hallmark of ARDS and is associated with the release of tissue-destructive immune effectors, such as matrix metalloproteinases (MMPs) and membrane-anchored metalloproteinase disintegrins (ADAMs). Here, we observed using intravital microscopy that Adam8-/- mice had impaired neutrophil transmigration. In mouse pneumonia models, both genetic deletion and pharmacologic inhibition of ADAM8 attenuated neutrophil infiltration and lung injury while improving bacterial containment. Unexpectedly, the alterations of neutrophil function were not attributable to impaired proteolysis but resulted from reduced intracellular interactions of ADAM8 with the actin-based motor molecule Myosin1f that suppressed neutrophil motility. In 2 ARDS cohorts, we analyzed lung fluid proteolytic signatures and identified that ADAM8 activity was positively correlated with disease severity. We propose that in acute inflammatory lung diseases such as pneumonia and ARDS, ADAM8 inhibition might allow fine-tuning of neutrophil responses for therapeutic gain.
Collapse
Affiliation(s)
- Catharina Conrad
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Daniela Yildiz
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Simon J. Cleary
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Lena Cook
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| | - Uwe Schlomann
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| | - Barry Panaretou
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Jessica L. Bowser
- Department of Pathology & Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Jiwen Li
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nathaniel K. Berg
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Ahmad Aljohmani
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - S. Farshid Moussavi-Harami
- Department of Pediatrics, Division of Pediatric Critical Care, University of California, San Francisco, San Francisco, California, USA
| | - Kristin M. Wang
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jennifer J. Tian
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Mélia Magnen
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Colin Valet
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Longhui Qiu
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jonathan P. Singer
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Wilhelm Bertrams
- Institute for Lung Research (iLung), Philipps-University, Marburg, Germany
| | - Susanne Herold
- Department of Internal Medicine II, University Medical Center Giessen and Marburg, Giessen, Germany
- Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Schmeck
- Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
- Pulmonary and Critical Care Medicine, University Medical Center Giessen and Marburg, Marburg, Germany
- German Center for Infectious Disease Research (DZIF), Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Zachary T. Ball
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Mark R. Looney
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jörg W. Bartsch
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| |
Collapse
|
27
|
Sinha P, Delucchi KL, Chen Y, Zhuo H, Abbott J, Wang C, Wickersham N, McNeil JB, Jauregui A, Ke S, Vessel K, Gomez A, Hendrickson CM, Kangelaris KN, Sarma A, Leligdowicz A, Liu KD, Matthay MA, Ware LB, Calfee CS. Latent class analysis-derived subphenotypes are generalisable to observational cohorts of acute respiratory distress syndrome: a prospective study. Thorax 2022; 77:13-21. [PMID: 34253679 PMCID: PMC8688287 DOI: 10.1136/thoraxjnl-2021-217158] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/01/2021] [Indexed: 01/03/2023]
Abstract
RATIONALE Using latent class analysis (LCA), two subphenotypes of acute respiratory distress syndrome (ARDS) have consistently been identified in five randomised controlled trials (RCTs), with distinct biological characteristics, divergent outcomes and differential treatment responses to randomised interventions. Their existence in unselected populations of ARDS remains unknown. We sought to identify subphenotypes in observational cohorts of ARDS using LCA. METHODS LCA was independently applied to patients with ARDS from two prospective observational cohorts of patients admitted to the intensive care unit, derived from the Validating Acute Lung Injury markers for Diagnosis (VALID) (n=624) and Early Assessment of Renal and Lung Injury (EARLI) (n=335) studies. Clinical and biological data were used as class-defining variables. To test for concordance with prior ARDS subphenotypes, the performance metrics of parsimonious classifier models (interleukin 8, bicarbonate, protein C and vasopressor-use), previously developed in RCTs, were evaluated in EARLI and VALID with LCA-derived subphenotypes as the gold-standard. RESULTS A 2-class model best fit the population in VALID (p=0.0010) and in EARLI (p<0.0001). Class 2 comprised 27% and 37% of the populations in VALID and EARLI, respectively. Consistent with the previously described 'hyperinflammatory' subphenotype, Class 2 was characterised by higher proinflammatory biomarkers, acidosis and increased shock and worse clinical outcomes. The similarities between these and prior RCT-derived subphenotypes were further substantiated by the performance of the parsimonious classifier models in both cohorts (area under the curves 0.92-0.94). The hyperinflammatory subphenotype was associated with increased prevalence of chronic liver disease and neutropenia and reduced incidence of chronic obstructive pulmonary disease. Measurement of novel biomarkers showed significantly higher levels of matrix metalloproteinase-8 and markers of endothelial injury in the hyperinflammatory subphenotype, whereas, matrix metalloproteinase-9 was significantly lower. CONCLUSION Previously described subphenotypes are generalisable to unselected populations of non-trauma ARDS.
Collapse
Affiliation(s)
- Pratik Sinha
- Department of Anesthesiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Kevin L Delucchi
- Department of Psychiatry, University of California San Francisco, San Francisco, California, USA
| | - Yue Chen
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Hanjing Zhuo
- Department of Anesthesiology, University of California San Francisco, San Francisco, California, USA
| | - Jason Abbott
- Department of Anesthesiology, University of California San Francisco, San Francisco, California, USA
| | - Chunxue Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nancy Wickersham
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - J Brennan McNeil
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alejandra Jauregui
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Serena Ke
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Kathryn Vessel
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Antonio Gomez
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Department of Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
| | - Carolyn M Hendrickson
- Department of Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
| | - Kirsten N Kangelaris
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- University of California San Francisco, San Francisco, California, USA
| | - Aartik Sarma
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, California, USA
| | | | - Kathleen D Liu
- Department of Anesthesiology, University of California San Francisco, San Francisco, California, USA
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, USA
| | - Lorraine B Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carolyn S Calfee
- Department of Anesthesiology, University of California San Francisco, San Francisco, California, USA
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, California, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
28
|
Jones TK, Reilly JP, Anderson BJ, Miano TA, Dunn TG, Weisman AR, Agyekum R, Feng R, Ittner CA, Shashaty MG, Meyer NJ. Elevated Plasma Levels of Matrix Metalloproteinase-3 and Tissue-Inhibitor of Matrix Metalloproteinases-1 Associate With Organ Dysfunction and Mortality in Sepsis. Shock 2022; 57:41-47. [PMID: 34265829 PMCID: PMC8663538 DOI: 10.1097/shk.0000000000001833] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Matrix Metalloproteinases (MMP) respond to tissue damage during sepsis. Higher plasma concentrations of MMPs and the tissue-inhibitor of matrix metalloproteinases (TIMP) have been reported in sepsis compared with healthy controls. The objective of this study was to examine if plasma levels of MMP-3, MMP-9, and TIMP-1 associate with mortality and organ dysfunction during sepsis. METHODS We conducted a prospective cohort study of critically ill patients with sepsis adjudicated per Sepsis-3 criteria at a tertiary academic medical center. We measured plasma concentrations of MMP-3, MMP-9, and TIMP-1 on intensive care unit admission. We phenotyped the subjects for shock, acute respiratory distress syndrome (ARDS), acute kidney injury (AKI), and mortality at 30 days. We used logistic regression to test the associations between the MMPs and TIMP-1 with shock, ARDS, AKI, and mortality. RESULTS Higher plasma TIMP-1 levels were associated with shock (odds ratio [OR] 1.51 per log increase [95% CI 1.25, 1.83]), ARDS (OR 1.24 [95% CI 1.05, 1.46]), AKI (OR 1.18 [95% CI 1.01, 1.38]), and mortality (OR 1.20 [95% CI 1.05, 1.46]. Higher plasma MMP-3 concentrations were associated with shock (OR 1.40 [95% CI 1.12, 1.75]) and mortality (OR 1.24 [95% CI 1.03, 1.48]) whereas MMP-9 levels were not associated with outcomes. Higher plasma TIMP-1 to MMP-3 ratios were associated with shock (OR 1.41 [95% CI 1.15, 1.72], P = 0.02). CONCLUSION Elevated plasma concentrations of TIMP-1 associate with organ dysfunction and mortality in sepsis. Higher plasma levels of MMP-3 associate with shock and mortality. Plasma MMP and TIMP-1 may warrant further investigation as emerging sepsis theragnostic biomarkers.
Collapse
Affiliation(s)
- Tiffanie K. Jones
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John P. Reilly
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brian J. Anderson
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd A. Miano
- Division of Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Thomas G. Dunn
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ariel R. Weisman
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Roseline Agyekum
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rui Feng
- Division of Biostatistics, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Caroline A.G. Ittner
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael G.S. Shashaty
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Kollaras V, Valsami G, Lambropoulou M, Konstandi O, Kostomistsopoulos N, Pikoulis E, Simopoulos C, Tsaroucha A. Effect of silibinin on the expression of MMP2, MMP3, MMP9 and TIMP2 in kidney and lung after hepatic ischemia/reperfusion injury in an experimental rat model. Acta Cir Bras 2021; 36:e360904. [PMID: 34755764 PMCID: PMC8580512 DOI: 10.1590/acb360904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/12/2021] [Indexed: 11/22/2022] Open
Abstract
PURPOSE The protective effect of silibinin on kidney and lung parenchyma during hepatic ischemia/reperfusion injury (IRI) is explored. METHODS Sixty-three Wistar rats were separated into three groups: sham; control (45 min IRI); and silibinin (200 μL silibinin administration after 45 min of ischemia and before reperfusion). Immunohistochemistry and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to evaluate the expression levels of MMP2, MMP3, MMP9, and TIMP2 on kidney and lung. RESULTS Comparing sham vs. control groups, confirmed that hepatic IRI increased both renal and lung MMP2, MMP3, MMP9 and TIMP2 expressions starting at 180 min (p<0.001). Comparison of the control vs. silibinin groups showed a statistically significant decrease in the expression levels of MMP2, MMP3, and MMP9 and increase of TIMP2 in kidney and lung parenchyma. The starting point of this decrease was at 120 min after reperfusion, both for kidney and lung parameters, and it was statistically significant at 240 min (p<0.001) for kidney, while silibinin showed a peak of lung protection at 180 min after hepatic reperfusion (p<0.001). CONCLUSIONS Hepatic IRI causes distant kidney and lung damage, while a statistically significant protective action, both on kidney and lung parenchyma, is conveyed by the intravenous administration of silibinin.
Collapse
|
30
|
Sinha P, Bos LD. Pathophysiology of the Acute Respiratory Distress Syndrome: Insights from Clinical Studies. Crit Care Clin 2021; 37:795-815. [PMID: 34548134 PMCID: PMC8149201 DOI: 10.1016/j.ccc.2021.05.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Pratik Sinha
- Division of Clinical and Translational Research, Department of Anesthesia, Washington University School of Medicine, 660 S. Euclid Avenue, Campus Box 8054, St Louis, MO 63110, USA.
| | - Lieuwe D Bos
- Department of Respiratory Medicine, Infection and Immunity, Amsterdam University Medical Center, AMC, Meibergdreef 9, Amsterdam 1105AZ, The Netherlands
| |
Collapse
|
31
|
Lin Y, Yang P. Phillygenin inhibits the inflammation and apoptosis of pulmonary epithelial cells by activating PPARγ signaling via downregulation of MMP8. Mol Med Rep 2021; 24:775. [PMID: 34490481 PMCID: PMC8441984 DOI: 10.3892/mmr.2021.12415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/10/2021] [Indexed: 01/01/2023] Open
Abstract
Acute lung injury (ALI) is often responsible for the high morbidity of critically ill patients. The present study aimed to investigate whether phillygenin (PHI) can inhibit inflammation and apoptosis of pulmonary epithelial cells by activating peroxisome proliferator-activated receptor γ (PPARγ) signaling. The in vitro model of ALI was established using lipopolysaccharide (LPS) and PHI was used to treat the LPS-induced cells. Cell viability was assessed using the MTT assay and the concentration levels of the inflammatory factors were detected by ELISA. Western blotting and reverse transcription-quantitative PCR were conducted to measure the expression levels of the inflammation- and apoptosis-associated proteins. The MMP8-overexpression plasmid was transfected into LPS-induced cells, which were treated with PHI treatment and the expression levels of PPARγ were detected via western blotting. PHI treatment suppressed the induction of inflammation and apoptosis of LPS-induced BEAS-2B cells. Furthermore, the expression levels of MMP8 in BEAS-2B cells induced by LPS were decreased following PHI treatment. Following transfection of the MMP8 overexpression plasmid into the LPS-induced BEAS-2B cells and subsequent treatment of these cells with PHI, the expression levels of PPARγ were decreased. In conclusion, it was shown that PHI inhibited the inflammation and apoptosis of pulmonary epithelial cells by activating PPARγ signaling via downregulating MMP8. These data may provide valuable information for future studies exploring the therapeutic effects of PHI for ALI.
Collapse
Affiliation(s)
- Yufeng Lin
- Department of Pediatrics, Gaolangang Hospital of Zhuhai People's Hospital, Zhuhai, Guangdong 519050, P.R. China
| | - Peng Yang
- Department of PICU, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
32
|
K 2P2.1 (TREK-1) potassium channel activation protects against hyperoxia-induced lung injury. Sci Rep 2020; 10:22011. [PMID: 33319831 PMCID: PMC7738539 DOI: 10.1038/s41598-020-78886-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
No targeted therapies exist to counteract Hyperoxia (HO)-induced Acute Lung Injury (HALI). We previously found that HO downregulates alveolar K2P2.1 (TREK-1) K+ channels, which results in worsening lung injury. This decrease in TREK-1 levels leaves a subset of channels amendable to pharmacological intervention. Therefore, we hypothesized that TREK-1 activation protects against HALI. We treated HO-exposed mice and primary alveolar epithelial cells (AECs) with the novel TREK-1 activators ML335 and BL1249, and quantified physiological, histological, and biochemical lung injury markers. We determined the effects of these drugs on epithelial TREK-1 currents, plasma membrane potential (Em), and intracellular Ca2+ (iCa) concentrations using fluorometric assays, and blocked voltage-gated Ca2+ channels (CaV) as a downstream mechanism of cytokine secretion. Once-daily, intra-tracheal injections of HO-exposed mice with ML335 or BL1249 improved lung compliance, histological lung injury scores, broncho-alveolar lavage protein levels and cell counts, and IL-6 and IP-10 concentrations. TREK-1 activation also decreased IL-6, IP-10, and CCL-2 secretion from primary AECs. Mechanistically, ML335 and BL1249 induced TREK-1 currents in AECs, counteracted HO-induced cell depolarization, and lowered iCa2+ concentrations. In addition, CCL-2 secretion was decreased after L-type CaV inhibition. Therefore, Em stabilization with TREK-1 activators may represent a novel approach to counteract HALI.
Collapse
|
33
|
Yehya N, Varisco BM, Thomas NJ, Wong HR, Christie JD, Feng R. Peripheral blood transcriptomic sub-phenotypes of pediatric acute respiratory distress syndrome. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:681. [PMID: 33287889 PMCID: PMC7720038 DOI: 10.1186/s13054-020-03410-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/24/2020] [Indexed: 02/18/2023]
Abstract
Background Acute respiratory distress syndrome (ARDS) is heterogeneous and may be amenable to sub-phenotyping to improve enrichment for trials. We aimed to identify subtypes of pediatric ARDS based on whole blood transcriptomics. Methods This was a prospective observational study of children with ARDS at the Children’s Hospital of Philadelphia (CHOP) between January 2018 and June 2019. We collected blood within 24 h of ARDS onset, generated expression profiles, and performed k-means clustering to identify sub-phenotypes. We tested the association between sub-phenotypes and PICU mortality and ventilator-free days at 28 days using multivariable logistic and competing risk regression, respectively. Results We enrolled 106 subjects, of whom 96 had usable samples. We identified three sub-phenotypes, dubbed CHOP ARDS Transcriptomic Subtypes (CATS) 1, 2, and 3. CATS-1 subjects (n = 31) demonstrated persistent hypoxemia, had ten subjects (32%) with immunocompromising conditions, and 32% mortality. CATS-2 subjects (n = 29) had more immunocompromising diagnoses (48%), rapidly resolving hypoxemia, and 24% mortality. CATS-3 subjects (n = 36) had the fewest comorbidities and also had rapidly resolving hypoxemia and 8% mortality. The CATS-3 subtype was associated with lower mortality (OR 0.18, 95% CI 0.04–0.86) and higher probability of extubation (subdistribution HR 2.39, 95% CI 1.32–4.32), relative to CATS-1 after adjustment for confounders. Conclusions We identified three sub-phenotypes of pediatric ARDS using whole blood transcriptomics. The sub-phenotypes had divergent clinical characteristics and prognoses. Further studies should validate these findings and investigate mechanisms underlying differences between sub-phenotypes.
Collapse
Affiliation(s)
- Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, 6040A Wood Building, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA. .,University of Pennsylvania, Philadelphia, PA, USA.
| | - Brian M Varisco
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Neal J Thomas
- Division of Pediatric Critical Care Medicine, Department of Pediatrics and Public Health Science, Penn State Hershey Children's Hospital, Hershey, PA, USA
| | - Hector R Wong
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jason D Christie
- Critical Care Division, Department of Medicine, Pulmonary, Allergy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Feng
- Department of Biostatistics, Center for Clinical Epidemiology and Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Endothelial Biomarkers Are Associated With Indirect Lung Injury in Sepsis-Associated Pediatric Acute Respiratory Distress Syndrome. Crit Care Explor 2020; 2:e0295. [PMID: 33299985 PMCID: PMC7721219 DOI: 10.1097/cce.0000000000000295] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Supplemental Digital Content is available in the text. Objectives: Acute respiratory distress syndrome occurring in the setting of direct versus indirect lung injury may reflect different pathobiologies amenable to different treatment strategies. We sought to test whether a panel of plasma biomarkers differed between children with sepsis-associated direct versus indirect acute respiratory distress syndrome. We hypothesized that a biomarker profile indicative of endothelial activation would be associated with indirect acute respiratory distress syndrome. Design: Observational cohort. Setting: Academic PICU. Subjects: Patients less than 18 years old with sepsis-associated direct (pneumonia, n = 52) or indirect (extrapulmonary sepsis, n = 46) acute respiratory distress syndrome. Interventions: None. Measurements and Main Results: Of 58 biomarkers examined, 33 differed by acute respiratory distress syndrome subtype. We used classification and regression tree methodology to examine associations between clinical and biochemical markers and acute respiratory distress syndrome subtype. The classification and regression tree model using only clinical variables (age, sex, race, oncologic comorbidity, and Pediatric Risk of Mortality-III score) performed worse than the classification and regression tree model using five clinical variables and 58 biomarkers. The best classification and regression tree model used only four endothelial biomarkers, including elevated angiopoietin-2/angiopoietin-1 ratio, vascular cell-adhesion molecule, and von Willebrand factor, to identify indirect acute respiratory distress syndrome. Test characteristics were 89% (80–97%) sensitivity, 80% (69–92%) specificity, positive predictive value 84% (74–93%), and negative predictive value 86% (76–96%). Conclusions: Indirect lung injury in children with acute respiratory distress syndrome is characterized by a biomarker profile indicative of endothelial activation, excess inflammation, and worse outcomes. A model using four biomarkers has the potential to be useful for more precisely identifying patients with acute respiratory distress syndrome whose pathobiology may respond to endothelial-targeted therapies in future trials.
Collapse
|
35
|
Abstract
OBJECTIVES To identify and compare serum and lower respiratory tract fluid biomarkers of lung injury using well-characterized mouse models of lung injury. To explore the relationship between these preclinical biomarkers and clinical outcomes in a discovery cohort of pediatric patients with acute respiratory failure from pneumonia. DESIGN Prospective, observational cohort study. SETTING A basic science laboratory and the PICU of a tertiary-care children's hospital. PATIENTS PICU patients intubated for respiratory failure from a suspected respiratory infection. INTERVENTIONS Prospective enrollment and collection of lower respiratory tract fluid samples. MEASUREMENTS AND MAIN RESULTS C57BL6/J mice were intranasally inoculated with escalating doses of influenza A virus or toll-like receptor agonists to simulate varying degrees of lung injury. Serum and bronchoalveolar lavage fluid were measured for the presence of cytokines using commercially available multiplex cytokine assays. Elevated levels of C-C motif chemokine ligand 7 at the peak of inflammation in both bronchoalveolar lavage fluid and serum correlated with lethality, with the bronchoalveolar lavage fluid ratio of C-C motif chemokine ligand 7:C-C motif chemokine ligand 22 providing the best prediction in the mouse models. These preclinical biomarkers were examined in the plasma and lower respiratory tract fluid of a discovery cohort of pediatric patients with acute respiratory failure from pneumonia. The primary clinical outcome measure was ventilator-free days, with secondary outcomes of pediatric acute respiratory distress syndrome severity and mortality. Elevation in peak lower respiratory tract fluid C-C motif chemokine ligand 7:C-C motif chemokine ligand 22 ratios demonstrated a significant negative correlation with ventilator-free days (r = -0.805; p < 0.02). CONCLUSIONS This study provides evidence that lung immune profiling via lower respiratory tract fluid cytokine analysis is feasible and may provide insight into clinical outcomes. Further validation of markers, including the C-C motif chemokine ligand 7:C-C motif chemokine ligand 22 ratio in this limited study, in a larger cohort of patients is necessary.
Collapse
|
36
|
Refining the Syndrome. Pediatr Crit Care Med 2020; 21:1094-1096. [PMID: 33278219 PMCID: PMC7725435 DOI: 10.1097/pcc.0000000000002570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
Makieieva N, Malakhova V, Vasylchenko Y, Biriukova M. Is Willebrand Factor Indicative of Chronic Inflammation in Children with Asthma? Turk Thorac J 2020; 21:362-366. [PMID: 33352089 DOI: 10.5152/turkthoracj.2019.19051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/02/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To improve our knowledge and to understand how the level of von Willebrand factor indicates the development of chronic inflammation in children with recurrent wheezing and asthma. MATERIAL AND METHODS It was a prospective cohort study. This study was conducted in children with recurrent wheezing and asthma who were referred to a children's hospital during 2017-2018. Patients were divided into 3 groups depending on the number of episodes of wheezing. Patients were examined for von Willebrand factor levels at admission and after treatment. Data analysis was performed with Statsofta Statistica Version 8 (Tulsa, OK). RESULTS WF1 levels in Group 2 and 3 children statistically significantly increased in comparison with the control group (p<0.001). WF2 levels remained elevated only in Group 3 patients (p<0.001). WF2 levels in Group 1 and 2 decreased to the indices of the control group (p>0.05). The WF2 significantly decreased after treatment in Group 2 children (p=0.0000, T=0) and Group 3 (p=0.0000, T=0). CONCLUSION levels of Willebrand factor indicate the presence of endothelial dysfunction. The level of Willebrand factor in the peak period of wheezing depends on the number of episodes of wheezing in history. Persistent high rates of Willebrand factor, even after the relief of clinical symptoms, indicates the present of chronic inflammation and can be regarded as the formation of asthma in children.
Collapse
Affiliation(s)
- Nataliia Makieieva
- Department of Pediatrics, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Valeriia Malakhova
- Department of Pediatrics, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Yuliia Vasylchenko
- Department of Pediatrics, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Maryna Biriukova
- Department of Pediatrics, Kharkiv National Medical University, Kharkiv, Ukraine
| |
Collapse
|
38
|
Geng P, Yu F, Tan D, Xu J, Yang Y, Xu M, Wang H, Ling B. Involvement of claudin-5 in H 2S-induced acute lung injury. J Toxicol Sci 2020; 45:293-304. [PMID: 32404561 DOI: 10.2131/jts.45.293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Acute exposure to hydrogen sulfide (H2S) can cause fatal acute lung injury (ALI). However, the mechanisms of H2S-induced ALI are still not fully understood. This study aims to investigate the role of the tight junction protein claudin-5 in H2S-induced ALI. In our study, Sprague-Dawley (SD) rats were exposed to H2S to establish the ALI model, and in parallel, human pulmonary microvascular endothelial cells (HPMECs) were incubated with NaHS (a H2S donor) to establish a cell model. Lung immunohistochemistry and electron microscopy assays were used to identify H2S-induced ALI, and the expression of claudin-5, p-AKT/t-AKT and p-FoxO1/t-FoxO1 was detected. Our results show that H2S promoted the formation of ALI by morphological investigation and decreased claudin-5 expression. Dexamethasone (Dex) could partly attenuate NaHS-mediated claudin-5 downregulation, and the protective effects of Dex could be partially blocked by LY294002, a PI3K/AKT/FoxO1 pathway antagonist. Moreover, as a consequence of the altered phosphorylation of AKT and FoxO1, a change in claudin-5 with the same trend was observed. Therefore, the tight junction protein claudin-5 might be considered a therapeutic target for the treatment of ALI induced by H2S and other hazardous gases.
Collapse
Affiliation(s)
- Ping Geng
- Department of Emergency Medicine, Northern Jiangsu People's Hospital, Yangzhou University College of Clinical Medicine, China
| | - Fen Yu
- Department of Emergency Medicine, Northern Jiangsu People's Hospital, Yangzhou University College of Clinical Medicine, China
| | - Dingyu Tan
- Department of Emergency Medicine, Northern Jiangsu People's Hospital, Yangzhou University College of Clinical Medicine, China
| | - Jiyang Xu
- Department of Emergency Medicine, Northern Jiangsu People's Hospital, Yangzhou University College of Clinical Medicine, China
| | - Yan Yang
- Department of Emergency Medicine, Northern Jiangsu People's Hospital, Yangzhou University College of Clinical Medicine, China
| | - Min Xu
- Department of Emergency Medicine, Northern Jiangsu People's Hospital, Yangzhou University College of Clinical Medicine, China
| | - Huihui Wang
- Department of Emergency Medicine, Northern Jiangsu People's Hospital, Yangzhou University College of Clinical Medicine, China
| | - Bingyu Ling
- Department of Emergency Medicine, Northern Jiangsu People's Hospital, Yangzhou University College of Clinical Medicine, China
| |
Collapse
|
39
|
Yang SC, Tsai YF, Pan YL, Hwang TL. Understanding the role of neutrophils in acute respiratory distress syndrome. Biomed J 2020; 44:439-446. [PMID: 33087299 PMCID: PMC7481802 DOI: 10.1016/j.bj.2020.09.001] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is difficult to treat and is associated with a high mortality rate. The most severe form of coronavirus disease 2019 (COVID-19) also leads to life-threatening ARDS. Neutrophil counts are positively correlated with disease severity in ARDS. Neutrophil activation not only plays a significant role in immune defense against infections, but also causes tissue damage and leads to inflammatory diseases. Activated neutrophils rapidly migrate to inflamed lung tissue, releasing toxic granular contents and generating neutrophil extracellular traps. In the last few decades, it has become apparent that neutrophils occupy a central role in ARDS pathology. In this review, we summarize the neutrophil inflammatory responses and their relationships to ARDS. According to the current literature, understanding the function of neutrophils may be helpful in the treatment of ARDS.
Collapse
Affiliation(s)
- Shun-Chin Yang
- Department of Anesthesiology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yung-Fong Tsai
- Department of Anesthesiology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yen-Lin Pan
- Department of Pharmacy, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan.
| |
Collapse
|
40
|
Kong M, Zhang WW, Sewell K, Gorman G, Kuo HC, Aban I, Ambalavanan N, Whitley RJ. Azithromycin Treatment vs Placebo in Children With Respiratory Syncytial Virus-Induced Respiratory Failure: A Phase 2 Randomized Clinical Trial. JAMA Netw Open 2020; 3:e203482. [PMID: 32324238 PMCID: PMC7180420 DOI: 10.1001/jamanetworkopen.2020.3482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPORTANCE Despite a high disease burden, there is no effective treatment for respiratory syncytial virus (RSV) infection. OBJECTIVES To determine whether administration of azithromycin (AZM) to children with RSV-induced respiratory failure is safe and to define the effect of AZM therapy on nasal matrix metalloproteinase 9 (MMP-9) levels. DESIGN, SETTING, AND PARTICIPANTS This randomized, double-blind, placebo-controlled phase 2 trial was conducted at a single tertiary pediatric intensive care unit from February 2016 to February 2019. The study included children with RSV infection who were admitted to the pediatric intensive care unit and required respiratory support via positive pressure ventilation (invasive and noninvasive). A total of 147 children were screened; 90 were excluded for not meeting inclusion criteria, having an absent legal guardian, lacking pharmacy support, or having a language barrier and 9 declined participation, resulting in 48 patients enrolled in the study. INTERVENTION Receipt of standard dose AZM (10 mg/kg/d), high-dose AZM (20 mg/kg/d), or a matching placebo of normal saline intravenously for 3 days. MAIN OUTCOMES AND MEASURES Nasal and endotracheal samples were collected at baseline as well as at 24 hours and 48 hours after start of treatment. The secondary outcome was to determine treatment effect on clinical outcome measures, including days of positive pressure ventilation and length of hospital stay. RESULTS A total of 48 patients were enrolled in the trial, with a median (range) age at randomization of 12 (1 to 125) months; 36 participants (75.0%) were younger than 2 years. Overall, 26 participants (54.2%) were boys, and 29 (60.4%) had a comorbidity. A total of 16 patients were randomized into each trial group (ie, placebo, standard-dose AZM, and high-dose AZM). Baseline demographic characteristics were comparable among the 3 groups. Both doses of AZM were safe, with no adverse events observed. No difference in nasal MMP-9 levels were observed between treatment groups. Among those who required mechanical ventilation and received high-dose AZM, endotracheal active and total MMP-9 levels were lower on day 3. Compared with baseline, active and total MMP-9 levels in endotracheal aspirates were 1.0 log lower in the high-dose AZM group (active MMP-9: 99.8% CI, -1.28 to -0.64; P < .001; total MMP-9: 99.8% CI, -1.37 to -0.57; P < .001). Patients who received high-dose AZM had fewer median (interquartile range) hospital days compared with those receiving the placebo (8 [6-14] days vs 11 [8-20] days; mean ratio estimate, 0.57; 95% CI, 0.38-0.87; P = .01). CONCLUSIONS AND RELEVANCE In this phase 2 randomized clinical trial, both doses of AZM were safe. While nasal MMP-9 levels were unchanged among treatment groups, endotracheal MMP-9 levels were lower among those who received high-dose AZM. The positive secondary clinical outcome, while exploratory, provides insight for end points in a multicenter randomized trial. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02707523.
Collapse
Affiliation(s)
- Michele Kong
- Department of Pediatrics, University of Alabama at Birmingham
| | - Wei Wei Zhang
- Department of Pediatrics, University of Alabama at Birmingham
| | - Kate Sewell
- Department of Pediatrics, University of Alabama at Birmingham
| | - Gregory Gorman
- McWhorter School of Pharmacy, Samford University, Birmingham, Alabama
| | - Hui-Chien Kuo
- Department of Pediatrics, University of Alabama at Birmingham
| | - Inmaculada Aban
- Department of Pediatrics, University of Alabama at Birmingham
| | | | | |
Collapse
|
41
|
Carlton EF, Flori HR. Biomarkers in pediatric acute respiratory distress syndrome. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:505. [PMID: 31728358 DOI: 10.21037/atm.2019.09.29] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Pediatric acute respiratory distress syndrome (PARDS) is a heterogenous process resulting in a severe acute lung injury. A single indicator does not exist for PARDS diagnosis. Rather, current diagnosis requires a combination of clinical and physiologic variables. Similarly, there is little ability to predict the path of disease, identify those at high risk of poor outcomes or target therapies specific to the underlying pathophysiology. Biomarkers, a measured indicator of a pathologic state or response to intervention, have been studied in PARDS due to their potential in diagnosis, prognostication and measurement of therapeutic response. Additionally, PARDS biomarkers show great promise in furthering our understanding of specific subgroups or endotypes in this highly variable disease, and thereby predict which patients may benefit and which may be harmed by PARDS specific therapies. In this chapter, we review the what, when, why and how of biomarkers in PARDS and discuss future directions in this quickly changing landscape.
Collapse
Affiliation(s)
- Erin F Carlton
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Heidi R Flori
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
42
|
Comprehensive Prognostication in Critically Ill Pediatric Hematopoietic Cell Transplant Patients: Results from Merging the Center for International Blood and Marrow Transplant Research (CIBMTR) and Virtual Pediatric Systems (VPS) Registries. Biol Blood Marrow Transplant 2019; 26:333-342. [PMID: 31563573 DOI: 10.1016/j.bbmt.2019.09.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 12/26/2022]
Abstract
Critically ill pediatric allogeneic hematopoietic cell transplant (HCT) patients may benefit from early and aggressive interventions aimed at reversing the progression of multiorgan dysfunction. Therefore, we evaluated 25 early risk factors for pediatric intensive care unit (PICU) mortality to improve mortality prognostication. We merged the Virtual Pediatric Systems and Center for International Blood and Marrow Transplant Research databases and analyzed 936 critically ill patients ≤21 years of age who had undergone allogeneic HCT and subsequently required PICU admission between January 1, 2009, and December 31, 2014. Of 1532 PICU admissions, the overall PICU mortality rate was 17.4% (95% confidence interval [CI], 15.6% to 19.4%) but was significantly higher for patients requiring mechanical ventilation (44.0%), renal replacement therapy (56.1%), or extracorporeal life support (77.8%). Mortality estimates increased significantly the longer that patients remained in the PICU. Of 25 HCT- and PICU-specific characteristics available at or near the time of PICU admission, moderate/severe pre-HCT renal injury, pre-HCT recipient cytomegalovirus seropositivity, <100-day interval between HCT and PICU admission, HCT for underlying acute myeloid leukemia, and greater admission organ dysfunction as approximated by the Pediatric Risk of Mortality 3 score were each independently associated with PICU mortality. A multivariable model using these components identified that patients in the top quartile of risk had 3 times greater mortality than other patients (35.1% versus 11.5%, P < .001, classification accuracy 75.2%; 95% CI, 73.0% to 77.4%). These data improve our working knowledge of the factors influencing the progression of critical illness in pediatric allogeneic HCT patients. Future investigation aimed at mitigating the effect of these risk factors is warranted.
Collapse
|
43
|
Aschner Y, Davidson JA. Early Plasma Matrix Metalloproteinase Profiles Offer New Insight into the Biology and Prognosis of Pediatric Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med 2019; 199:134-136. [PMID: 30160977 DOI: 10.1164/rccm.201808-1500ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Yael Aschner
- 1 Department of Medicine University of Colorado Aurora, Colorado and
| | - Jesse A Davidson
- 2 Department of Pediatrics University of Colorado/Children's Hospital Colorado| Aurora, Colorado
| |
Collapse
|
44
|
Wan H, Xie T, Xu Q, Hu X, Xing S, Yang H, Gao Y, He Z. Thy-1 depletion and integrin β3 upregulation-mediated PI3K-Akt-mTOR pathway activation inhibits lung fibroblast autophagy in lipopolysaccharide-induced pulmonary fibrosis. J Transl Med 2019; 99:1636-1649. [PMID: 31249375 PMCID: PMC7102294 DOI: 10.1038/s41374-019-0281-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/13/2019] [Accepted: 05/02/2019] [Indexed: 12/24/2022] Open
Abstract
Lipopolysaccharide (LPS)-induced autophagy inhibition in lung fibroblasts is closely associated with the activation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K-Akt-mTOR) pathway. However, the underlying mechanism remains unknown. In this study, we demonstrated that LPS activated the PI3K-Akt-mTOR pathway and inhibited lung fibroblast autophagy by depleting thymocyte differentiation antigen-1 (Thy-1) and upregulating integrin β3 (Itgb3). Challenge of the human lung fibroblast MRC-5 cell line with LPS resulted in significant upregulation of integrin β3, activation of the PI3K-Akt-mTOR pathway and inhibition of autophagy, which could be abolished by integrin β3 silencing by specific shRNA or treatment with the integrin β3 inhibitor cilengitide. Meanwhile, LPS could inhibit Thy-1 expression accompanied with PI3K-Akt-mTOR pathway activation and lung fibroblast autophagy inhibition; these effects could be prevented by Thy-1 overexpression. Meanwhile, Thy-1 downregulation with Thy-1 shRNA could mimic the effects of LPS, inducing the activation of PI3K-Akt-mTOR pathway and inhibiting lung fibroblast autophagy. Furthermore, protein immunoprecipitation analysis demonstrated that LPS reduced the binding of Thy-1 to integrin β3. Thy-1 downregulation, integrin β3 upregulation and autophagy inhibition were also detected in a mouse model of LPS-induced pulmonary fibrosis, which could be prohibited by intratracheal injection of Thy-1 overexpressing adeno-associated virus (AAV) or intraperitoneal injection of the integrin β3 inhibitor cilengitide. In conclusion, this study demonstrated that Thy-1 depletion and integrin β3 upregulation are involved in LPS-induced pulmonary fibrosis, and may serve as potential therapeutic targets for pulmonary fibrosis.
Collapse
Affiliation(s)
- Hanxi Wan
- 0000 0004 0368 8293grid.16821.3cDepartment of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 200127 Shanghai, China
| | - Tingting Xie
- 0000 0004 0368 8293grid.16821.3cDepartment of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 200127 Shanghai, China
| | - Qiaoyi Xu
- 0000 0004 0368 8293grid.16821.3cDepartment of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 200127 Shanghai, China
| | - Xiaoting Hu
- 0000 0004 0368 8293grid.16821.3cDepartment of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 200127 Shanghai, China
| | - Shunpeng Xing
- 0000 0004 0368 8293grid.16821.3cDepartment of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 200127 Shanghai, China
| | - Hao Yang
- 0000000123704535grid.24516.34Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433 Shanghai, China
| | - Yuan Gao
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 200127, Shanghai, China.
| | - Zhengyu He
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 200127, Shanghai, China.
| |
Collapse
|