Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L232-L249, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38860845

ABSTRACT

COVID-19 syndrome is characterized by acute lung injury, hypoxemic respiratory failure, and high mortality. Alveolar type 2 (AT2) cells are essential for gas exchange, repair, and regeneration of distal lung epithelium. We have shown that the causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and other members of the Ɵ-coronavirus genus induce an endoplasmic reticulum (ER) stress response in vitro; however, the consequences for host AT2 cell function in vivo are less understood. To study this, two murine models of coronavirus infection were used-mouse hepatitis virus-1 (MHV-1) in A/J mice and a mouse-adapted SARS-CoV-2 strain. MHV-1-infected mice exhibited dose-dependent weight loss with histological evidence of distal lung injury accompanied by elevated bronchoalveolar lavage fluid (BALF) cell counts and total protein. AT2 cells showed evidence of both viral infection and increased BIP/GRP78 expression, consistent with activation of the unfolded protein response (UPR). The AT2 UPR included increased inositol-requiring enzyme 1α (IRE1α) signaling and a biphasic response in PKR-like ER kinase (PERK) signaling accompanied by marked reductions in AT2 and BALF surfactant protein (SP-B and SP-C) content, increases in surfactant surface tension, and emergence of a reprogrammed epithelial cell population (Krt8+ and Cldn4+). The loss of a homeostatic AT2 cell state was attenuated by treatment with the IRE1α inhibitor OPK-711. As a proof-of-concept, C57BL6 mice infected with mouse-adapted SARS-CoV-2 demonstrated similar lung injury and evidence of disrupted surfactant homeostasis. We conclude that lung injury from Ɵ-coronavirus infection results from an aberrant host response, activating multiple AT2 UPR stress pathways, altering surfactant metabolism/function, and changing AT2 cell state, offering a mechanistic link between SARS-CoV-2 infection, AT2 cell biology, and acute respiratory failure.NEW & NOTEWORTHY COVID-19 syndrome is characterized by hypoxemic respiratory failure and high mortality. In this report, we use two murine models to show that Ɵ-coronavirus infection produces acute lung injury, which results from an aberrant host response, activating multiple epithelial endoplasmic reticular stress pathways, disrupting pulmonary surfactant metabolism and function, and forcing emergence of an aberrant epithelial transition state. Our results offer a mechanistic link between SARS-CoV-2 infection, AT2 cell biology, and respiratory failure.


Subject(s)
COVID-19 , Endoplasmic Reticulum Stress , Endoribonucleases , Homeostasis , Murine hepatitis virus , SARS-CoV-2 , Animals , Mice , COVID-19/metabolism , COVID-19/pathology , COVID-19/virology , COVID-19/complications , Murine hepatitis virus/pathogenicity , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Alveolar Epithelial Cells/pathology , Endoplasmic Reticulum Chaperone BiP , Coronavirus Infections/metabolism , Coronavirus Infections/pathology , Coronavirus Infections/virology , Coronavirus Infections/complications , Pulmonary Surfactants/metabolism , Unfolded Protein Response , Betacoronavirus , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/virology , Respiratory Insufficiency/pathology , Disease Models, Animal , eIF-2 Kinase/metabolism , Humans
2.
J Immunol ; 208(7): 1525-1533, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35288471

ABSTRACT

Severe asthma is characterized by steroid insensitivity and poor symptom control and is responsible for most asthma-related hospital costs. Therapeutic options remain limited, in part due to limited understanding of mechanisms driving severe asthma. Increased arginine methylation, catalyzed by protein arginine methyltransferases (PRMTs), is increased in human asthmatic lungs. In this study, we show that PRMT5 drives allergic airway inflammation in a mouse model reproducing multiple aspects of human severe asthma. We find that PRMT5 is required in CD4+ T cells for chronic steroid-insensitive severe lung inflammation, with selective T cell deletion of PRMT5 robustly suppressing eosinophilic and neutrophilic lung inflammation, pathology, airway remodeling, and hyperresponsiveness. Mechanistically, we observed high pulmonary sterol metabolic activity, retinoic acid-related orphan receptor ƎĀ³t (RORƎĀ³t), and Th17 responses, with PRMT5-dependent increases in RORƎĀ³t's agonist desmosterol. Our work demonstrates that T cell PRMT5 drives severe allergic lung inflammation and has potential implications for the pathogenesis and therapeutic targeting of severe asthma.


Subject(s)
Asthma , Hypersensitivity , Animals , Asthma/metabolism , Granulocytes/metabolism , Hypersensitivity/metabolism , Inflammation/metabolism , Mice , Th17 Cells/metabolism
3.
Nat Immunol ; 12(3): 222-30, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21151103

ABSTRACT

Autophagy, a cellular process for organelle and protein turnover, regulates innate immune responses. Here we demonstrate that depletion of the autophagic proteins LC3B and beclin 1 enhanced the activation of caspase-1 and secretion of interleukin 1Ɵ (IL-1Ɵ) and IL-18. Depletion of autophagic proteins promoted the accumulation of dysfunctional mitochondria and cytosolic translocation of mitochondrial DNA (mtDNA) in response to lipopolysaccharide (LPS) and ATP in macrophages. Release of mtDNA into the cytosol depended on the NALP3 inflammasome and mitochondrial reactive oxygen species (ROS). Cytosolic mtDNA contributed to the secretion of IL-1Ɵ and IL-18 in response to LPS and ATP. LC3B-deficient mice produced more caspase-1-dependent cytokines in two sepsis models and were susceptible to LPS-induced mortality. Our study suggests that autophagic proteins regulate NALP3-dependent inflammation by preserving mitochondrial integrity.


Subject(s)
Autophagy , Carrier Proteins/immunology , DNA, Mitochondrial , Immunity, Innate , Inflammasomes/immunology , Animals , Caspase 1/immunology , Flow Cytometry , Mice , NLR Family, Pyrin Domain-Containing 3 Protein
4.
Toxicol Appl Pharmacol ; 459: 116341, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36502870

ABSTRACT

Asthma is a chronic inflammatory airway disease characterized by acute exacerbations triggered by inhaled allergens, respiratory infections, or air pollution. Ozone (O3), a major component of air pollution, can damage the lung epithelium in healthy individuals. Despite this association, little is known about the effects of O3 and its impact on chronic lung disease. Epidemiological data have demonstrated that elevations in ambient O3 are associated with increased asthma exacerbations. To identify mechanisms by which O3 exposure leads to asthma exacerbations, we developed a two-hit mouse model where mice were sensitized and challenged with three common allergens (dust mite, ragweed and Aspergillus fumigates, DRA) to induce allergic inflammation prior to exposure to O3 (DRAO3). Changes in lung physiology, inflammatory cells, and inflammation were measured. Exposure to O3 following DRA significantly increased airway hyperreactivity (AHR), which was independent of TLR4. DRA exposure resulted in increased BAL eosinophilia while O3 exposure resulted in neutrophilia. Additionally, O3 exposure following DRA blunted anti-inflammatory and antioxidant responses. Finally, there were significantly less monocytes and innate lymphoid type 2 cells (ILC2s) in the dual challenged DRA-O3 group suggesting that the lack of these immune cells may influence O3-induced AHR in the setting of allergic inflammation. In summary, we developed a mouse model that mirrors some aspects of the clinical course of asthma exacerbations due to air pollution and identified that O3 exposure in the asthmatic lung leads to impaired endogenous anti-inflammatory and antioxidant responses and alterations inflammatory cell populations.


Subject(s)
Asthma , Eosinophilia , Ozone , Mice , Animals , Ozone/toxicity , Immunity, Innate , Antioxidants/pharmacology , Lymphocytes , Asthma/chemically induced , Lung , Inflammation , Allergens/toxicity , Disease Models, Animal
5.
Pulm Pharmacol Ther ; 79: 102196, 2023 04.
Article in English | MEDLINE | ID: mdl-36682407

ABSTRACT

The acute respiratory distress syndrome (ARDS) is a life-threatening condition that causes respiratory failure. Despite numerous clinical trials, there are no molecularly targeted pharmacologic therapies to prevent or treat ARDS. Drug delivery during ARDS is challenging due to the heterogenous nature of lung injury and occlusion of lung units by edema fluid and inflammation. Pulmonary drug delivery during ARDS offers several potential advantages including limiting the off-target and off-organ effects and directly targeting the damaged and inflamed lung regions. In this review we summarize recent ARDS clinical trials using both systemic and pulmonary drug delivery. We then discuss the advantages of pulmonary drug delivery and potential challenges to its implementation. Finally, we discuss the use of nanoparticle drug delivery and surfactant-based drug carriers as potential strategies for delivering therapeutics to the injured lung in ARDS.


Subject(s)
Pulmonary Surfactants , Respiratory Distress Syndrome , Humans , Lung , Respiratory Distress Syndrome/drug therapy , Drug Delivery Systems , Pulmonary Surfactants/therapeutic use , Drug Carriers
6.
Am J Respir Cell Mol Biol ; 66(2): e1-e14, 2022 02.
Article in English | MEDLINE | ID: mdl-35103557

ABSTRACT

Advancements in methods, technology, and our understanding of the pathobiology of lung injury have created the need to update the definition of experimental acute lung injury (ALI). We queried 50 participants with expertise in ALI and acute respiratory distress syndrome using a Delphi method composed of a series of electronic surveys and a virtual workshop. We propose that ALI presents as a "multidimensional entity" characterized by four "domains" that reflect the key pathophysiologic features and underlying biology of human acute respiratory distress syndrome. These domains are 1) histological evidence of tissue injury, 2) alteration of the alveolar-capillary barrier, 3) presence of an inflammatory response, and 4) physiologic dysfunction. For each domain, we present "relevant measurements," defined as those proposed by at least 30% of respondents. We propose that experimental ALI encompasses a continuum of models ranging from those focusing on gaining specific mechanistic insights to those primarily concerned with preclinical testing of novel therapeutics or interventions. We suggest that mechanistic studies may justifiably focus on a single domain of lung injury, but models must document alterations of at least three of the four domains to qualify as "experimental ALI." Finally, we propose that a time criterion defining "acute" in ALI remains relevant, but the actual time may vary based on the specific model and the aspect of injury being modeled. The continuum concept of ALI increases the flexibility and applicability of the definition to multiple models while increasing the likelihood of translating preclinical findings to critically ill patients.


Subject(s)
Acute Lung Injury/pathology , Inflammation/physiopathology , Research Report/trends , Acute Lung Injury/immunology , Animals
7.
J Intensive Care Med ; 37(3): 385-392, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33779386

ABSTRACT

BACKGROUND: Acute Respiratory Distress Syndrome (ARDS) is a frequent cause of respiratory failure in intensive care unit (ICU) patients and results in significant morbidity and mortality. ARDS often develops as a result of a local or systemic inflammatory insult. Cancer can lead to systemic inflammation but whether cancer is an independent risk factor for developing ARDS is unknown. We hypothesized that critically ill cancer patients admitted to the ICU were at increased risk for the diagnosis of ARDS. METHODS: Retrospective cohort study of critically ill patients admitted between July 2017 and December 2018 at an academic medical center in Columbus, Ohio. The primary outcome was the association of patients with malignancy and the diagnosis of ARDS in a multivariable logistic regression model with covariables selected a priori informed through the construction of a directed acyclic graph. RESULTS: 412 ARDS cases were identified with 166 of those patients having active cancer. There was an association between cancer and ARDS, with an odds ratio (OR) of 1.55 (95% CI 1.26-1.92, P < 0.001). When adjusted for our pre-specified confounding variables, the association remained statistically significant (OR 1.57, 95% CI 1.15-2.13, P = 0.004). In an unadjusted pre-specified subgroup analysis, hematologic malignancy (OR 1.81, 95% CI 1.30-2.53, P < 0.001) was associated with increased odds of developing ARDS while non-metastatic solid tumors (OR 0.51, 95% CI 0.31-0.85, P = 0.01) had statistically significant negative association. Cancer patients with ARDS had a significantly higher ICU (70.5% vs 39.8%, P < 0.001) and hospital (72.9% vs 40.7%, P < 0.001) mortality compared to ARDS patients without active malignancy. CONCLUSION: In this single center retrospective cohort study, cancer was found to be an independent risk factor for the diagnosis of ARDS in critically ill patients. To our knowledge, we are the first report an independent association between cancer and ARDS in critically ill patients.


Subject(s)
Neoplasms , Respiratory Distress Syndrome , Critical Illness , Humans , Intensive Care Units , Neoplasms/complications , Respiratory Distress Syndrome/epidemiology , Respiratory Distress Syndrome/etiology , Retrospective Studies , Risk Factors
8.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L921-L930, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32159972

ABSTRACT

The incidence of asthma has increased from 5.5% to near 8% of the population, which is a major health concern. The hallmarks of asthma include eosinophilic airway inflammation that is associated with chronic airway remodeling. Allergic airway inflammation is characterized by a complex interplay of resident and inflammatory cells. MicroRNAs (miRNAs) are small noncoding RNAs that function as posttranscriptional modulators of gene expression. However, the role of miRNAs, specifically miR-451, in the regulation of allergic airway inflammation is unexplored. Our previous findings showed that oxidant stress regulates miR-451 gene expression in macrophages during an inflammatory process. In this paper, we examined the role of miR-451 in regulating macrophage phenotype using an experimental poly-allergenic murine model of allergic airway inflammation. We found that miR-451 contributes to the allergic induction of CCL17 in the lung and plays a key role in proasthmatic macrophage activation. Remarkably, administration of a Sirtuin 2 (Sirt2) inhibitor diminished alternate macrophage activation and markedly abrogated triple-allergen [dust mite, ragweed, Aspergillus fumigatus (DRA)]-induced lung inflammation. These data demonstrate a role for miR-451 in modulating allergic inflammation by influencing allergen-mediated macrophages phenotype.


Subject(s)
Asthma/genetics , Macrophages, Alveolar/immunology , MicroRNAs/genetics , Pneumonia/genetics , Sirtuin 2/genetics , Allergens/administration & dosage , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Plant/administration & dosage , Aspergillus/chemistry , Aspergillus/immunology , Asthma/chemically induced , Asthma/pathology , Asthma/therapy , Chemokine CCL17/genetics , Chemokine CCL17/immunology , Disease Models, Animal , Fungi/chemistry , Fungi/immunology , Furans/pharmacology , Gene Expression Regulation , Lung/drug effects , Lung/immunology , Lung/pathology , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/immunology , Plant Extracts/administration & dosage , Pneumonia/chemically induced , Pneumonia/pathology , Pneumonia/therapy , Pyroglyphidae/chemistry , Pyroglyphidae/immunology , Quinolines/pharmacology , Signal Transduction , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/immunology
9.
Respir Res ; 20(1): 218, 2019 Oct 12.
Article in English | MEDLINE | ID: mdl-31606045

ABSTRACT

BACKGROUND: Respiratory pathology is a major driver of mortality in the intensive care unit (ICU), even in the absence of a primary respiratory diagnosis. Prior work has demonstrated that a visual scoring system applied to chest radiographs (CXR) is associated with adverse outcomes in ICU patients with Acute Respiratory Distress Syndrome (ARDS). We hypothesized that a simple, semi-quantitative CXR score would be associated with clinical outcomes for the general ICU population, regardless of underlying diagnosis. METHODS: All individuals enrolled in the Registry of Critical Illness at Brigham and Women's Hospital between June 2008 and August 2018 who had a CXR within 24 h of admission were included. Each patient's CXR was assigned an opacification score of 0-4 in each of four quadrants with the total score being the sum of all four quadrants. Multivariable negative binomial, logistic, and Cox regression, adjusted for age, sex, race, immunosuppression, a history of chronic obstructive pulmonary disease, a history of congestive heart failure, and APACHE II scores, were used to assess the total score's association with ICU length of stay (LOS), duration of mechanical ventilation, in-hospital mortality, 60-day mortality, and overall mortality, respectively. RESULTS: A total of 560 patients were included. Higher CXR scores were associated with increased mortality; for every one-point increase in score, in-hospital mortality increased 10% (OR 1.10, CI 1.05-1.16, p < 0.001) and 60-day mortality increased by 12% (OR 1.12, CI 1.07-1.17, p < 0.001). CXR scores were also independently associated with both ICU length of stay (rate ratio 1.06, CI 1.04-1.07, p < 0.001) and duration of mechanical ventilation (rate ratio 1.05, CI 1.02-1.07, p < 0.001). CONCLUSIONS: Higher values on a simple visual score of a patient's CXR on admission to the medical ICU are associated with increased in-hospital mortality, 60-day mortality, overall mortality, length of ICU stay, and duration of mechanical ventilation.


Subject(s)
Critical Illness , Respiratory Distress Syndrome/diagnostic imaging , Thorax/diagnostic imaging , APACHE , Adult , Aged , Biomarkers/blood , Female , Hospital Mortality , Humans , Length of Stay , Lung/diagnostic imaging , Male , Middle Aged , Organ Size , Respiration, Artificial , Respiratory Distress Syndrome/mortality , Respiratory Distress Syndrome/therapy , Retrospective Studies , Treatment Outcome
10.
Respir Res ; 20(1): 15, 2019 Jan 21.
Article in English | MEDLINE | ID: mdl-30665420

ABSTRACT

BACKGROUND: The acute respiratory distress syndrome (ARDS) is characterized by the acute onset of hypoxemia and bilateral lung infiltrates in response to an inciting event, and is associated with high morbidity and mortality. Patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT) are at increased risk for ARDS. We hypothesized that HSCT patients with ARDS would have a unique transcriptomic profile identifiable in peripheral blood compared to those that did not undergo HSCT. METHODS: We isolated RNA from banked peripheral blood samples from a biorepository of critically ill ICU patients. RNA-SeqĀ was performed on 11 patients with ARDS (5 that had undergone HSCT and 6 that had not) and 12 patients with sepsis without ARDS (5 that that had undergone HCST and 7 that had not). RESULTS: We identified 687 differentially expressed genes between ARDS and ARDS-HSCT (adjusted p-value < 0.01), including IFI44L, OAS3, LY6E, and SPATS2L that had increased expression in ARDS vs. ARDS-HSCT; these genes were not differentially expressed in sepsis vs sepsis-HSCT. Gene ontology enrichment analysis revealed that many differentially expressed genes were related to response to type I interferon. CONCLUSIONS: Our findings reveal significant differences in whole blood transcriptomic profiles of patients with non-HSCT ARDS compared to ARDS-HSCT patients and point toward different immune responses underlying ARDS and ARDS-HSCT that contribute to lung injury.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/therapy , Sequence Analysis, RNA/methods , Transcriptome/genetics , Adult , Female , Hematopoietic Stem Cell Transplantation/trends , Humans , Male , Middle Aged , Registries , Respiratory Distress Syndrome/blood , Sequence Analysis, RNA/trends
11.
Allergy ; 74(3): 535-548, 2019 03.
Article in English | MEDLINE | ID: mdl-30288751

ABSTRACT

BACKGROUND: The pathogenesis of asthma and airway obstruction is the result of an abnormal response to different environmental exposures. The scientific premise of our study was based on the finding that FoxO1 expression is increased in lung macrophages of mice after allergen exposure and human asthmatic patients. Macrophages are capable of switching from one functional phenotype to another, and it is important to understand the mechanisms involved in the transformation of macrophages and how their cellular function affects the peribronchial stromal microenvironment. METHODS: We employed a murine asthma model, in which mice were treated by intranasal insufflation with allergens for 2-8Ā weeks. We used both a pharmacologic approach using a highly specific FoxO1 inhibitor and genetic approaches using FoxO1 knockout mice (FoxO1fl/fl LysMcre). Cytokine level in biological fluids was measured by ELISA and the expression of encoding molecules by NanoString assay and qRT-PCR. RESULTS: We show that the levels of FoxO1 gene are significantly elevated in the airway macrophages of patients with mild asthma in response to subsegmental bronchial allergen challenge. Transcription factor FoxO1 regulates a pro-asthmatic phenotype of lung macrophages that is involved in the development and progression of chronic allergic airway disease. We have shown that inhibition of FoxO1 induced phenotypic conversion of lung macrophages and downregulates pro-asthmatic and pro-fibrotic gene expression by macrophages, which contribute to airway inflammation and airway remodeling in allergic asthma. CONCLUSION: Targeting FoxO1 with its downstream regulator IRF4 is a novel therapeutic target for controlling allergic inflammation and potentially reversing fibrotic airway remodeling.


Subject(s)
Asthma/etiology , Asthma/metabolism , Forkhead Box Protein O1/genetics , Gene Expression Regulation, Neoplastic , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Adoptive Transfer , Allergens/immunology , Animals , Asthma/diagnosis , Asthma/therapy , Bronchial Provocation Tests , Bronchoscopy , Disease Models, Animal , Female , Flow Cytometry , Forkhead Box Protein O1/metabolism , Humans , Mice , Th2 Cells/immunology , Th2 Cells/metabolism
12.
Am J Respir Cell Mol Biol ; 56(3): 300-309, 2017 03.
Article in English | MEDLINE | ID: mdl-27636016

ABSTRACT

Oxidative stress resulting from inflammatory responses that occur during acute lung injury and sepsis can initiate changes in mitochondrial function. Autophagy regulates cellular processes in the setting of acute lung injury, sepsis, and oxidative stress by modulating the immune response and facilitating turnover of damaged cellular components. We have shown that mesenchymal stromal cells (MSCs) improve survival in murine models of sepsis by also regulating the immune response. However, the effect of autophagy on MSCs and MSC mitochondrial function during oxidative stress is unknown. This study investigated the effect of depletion of autophagic protein microtubule-associated protein 1 light chain 3B (LC3B) and beclin 1 (BECN1) on the response of MSCs to oxidative stress. MSCs were isolated from wild-type (WT) and LC3B-/- or Becn1+/- mice. MSCs from the LC3B-/- and Becn1+/- animals had increased susceptibility to oxidative stress-induced cell death as compared with WT MSCs. The MSCs depleted of autophagic proteins also had impaired mitochondrial function (decreased intracellular ATP, reduced mitochondrial membrane potential, and increased mitochondrial reactive oxygen species production) under oxidative stress as compared with WT MSCs. In WT MSCs, carbon monoxide (CO) preconditioning enhanced autophagy and mitophagy, and rescued the cells from oxidative stress-induced death. CO preconditioning was not able to rescue the decreased survival of MSCs from the LC3B-/- and Becn1+/- animals, further supporting the tenet that CO exerts its cytoprotective effects via the autophagy pathway.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mitochondria/metabolism , Oxidative Stress , Adenosine Triphosphate/metabolism , Animals , Antioxidants/metabolism , Autophagy/drug effects , Carbon Monoxide/pharmacology , Cells, Cultured , Intracellular Space/metabolism , Membrane Potential, Mitochondrial/drug effects , Mesenchymal Stem Cells/drug effects , Mice , Mitochondria/drug effects , Mitophagy/drug effects , Oxidative Stress/drug effects , Phenotype
13.
Am J Physiol Renal Physiol ; 312(4): F654-F660, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28365585

ABSTRACT

In critical illness, such as sepsis or the acute respiratory distress syndrome, acute kidney injury (AKI) is common and associated with increased morbidity and mortality. Mechanical ventilation in critical illnesses is also a risk factor for AKI, but it is potentially modifiable. Injurious ventilation strategies may lead to the systemic release of inflammatory mediators from the lung due to ventilator induced lung injury (VILI). The systemic consequences of VILI are difficult to differentiate clinically from other systemic inflammatory syndromes, such as sepsis. The purpose of this study was to identify unique changes in the expression of inflammatory mediators in kidney tissue in response to VILI compared with systemic sepsis to gain insight into direct effects of VILI on the kidney. Four groups of mice were compared-mice with sepsis from cecal ligation and puncture (CLP), mice subjected to injurious mechanical ventilation with high tidal volumes (VILI), mice exposed to CLP followed by VILI (CLP+VILI), and sham controls. Protein expression of common inflammatory mediators in kidneys was analyzed using a proteome array and confirmed by Western blot analysis or ELISA. VEGF and VCAM-1 were found to be significantly elevated in kidneys from VILI mice compared with sham and CLP. Angiopoietin-2 was significantly increased in CLP+VILI compared with CLP alone and was also correlated with higher levels of AKI biomarker, neutrophil gelatinase-associated lipocalin. These results suggest that VILI alters the renal expression of VEGF, VCAM-1, and angiopoietin-2, and these proteins warrant further investigation as potential biomarkers and therapeutic targets.


Subject(s)
Angiopoietin-2/metabolism , Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Kidney/blood supply , Respiration, Artificial/adverse effects , Sepsis/metabolism , Ventilator-Induced Lung Injury/metabolism , Animals , Biomarkers/metabolism , Disease Models, Animal , Male , Mice, Inbred C57BL , Proteomics/methods , Sepsis/microbiology , Signal Transduction , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Ventilator-Induced Lung Injury/etiology
17.
Anesthesiology ; 123(2): 377-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26068207

ABSTRACT

BACKGROUND: Isoflurane may be protective in preclinical models of lung injury, but its use in patients with lung injury remains controversial and the mechanism of its protective effects remains unclear. The authors hypothesized that this protection is mediated at the level of alveolar tight junctions and investigated the possibility in a two-hit model of lung injury that mirrors human acute respiratory distress syndrome. METHODS: Wild-type mice were treated with isoflurane 1 h after exposure to nebulized endotoxin (n = 8) or saline control (n = 9) and then allowed to recover for 24 h before mechanical ventilation (MV; tidal volume, 15 ml/kg, 2 h) producing ventilator-induced lung injury. Mouse lung epithelial cells were similarly treated with isoflurane 1 h after exposure to lipopolysaccharide. Cells were cyclically stretched the following day to mirror the MV protocol used in vivo. RESULTS: Mice treated with isoflurane following exposure to inhaled endotoxin and before MV exhibited significantly less physiologic lung dysfunction. These effects appeared to be mediated by decreased vascular leak, but not altered inflammatory indices. Mouse lung epithelial cells treated with lipopolysaccharide and cyclic stretch and lungs harvested from mice after treatment with lipopolysaccharide and MV had decreased levels of a key tight junction protein (i.e., zona occludens 1) that was rescued by isoflurane treatment. CONCLUSIONS: Isoflurane rescued lung injury induced by a two-hit model of endotoxin exposure followed by MV by maintaining the integrity of the alveolar-capillary barrier possibly by modulating the expression of a key tight junction protein.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Anesthetics, Inhalation/administration & dosage , Isoflurane/administration & dosage , Tight Junctions/metabolism , Acute Lung Injury/pathology , Animals , Cell Line, Transformed , Male , Mice , Mice, Inbred C57BL , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Tight Junctions/drug effects
18.
Res Sq ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38562870

ABSTRACT

The lung is a dynamic mechanical organ and several pulmonary disorders are characterized by heterogeneous changes in the lung's local mechanical properties (i.e. stiffness). These alterations lead to abnormal lung tissue deformation (i.e. strain) which have been shown to promote disease progression. Although heterogenous mechanical properties may be important biomarkers of disease, there is currently no non-invasive way to measure these properties for clinical diagnostic purposes. In this study, we use a magnetic resonance elastography technique to measure heterogenous distributions of the lung's shear stiffness in healthy adults and in people with Cystic Fibrosis. Additionally, computational finite element models which directly incorporate the measured heterogenous mechanical properties were developed to assess the effects on lung tissue deformation. Results indicate that consolidated lung regions in people with Cystic Fibrosis exhibited increased shear stiffness and reduced spatial heterogeneity compared to surrounding non-consolidated regions. Accounting for heterogenous lung stiffness in healthy adults did not change the globally averaged strain magnitude obtained in computational models. However, computational models that used heterogenous stiffness measurements predicted significantly more variability in local strain and higher spatial strain gradients. Finally, computational models predicted lower strain variability and spatial strain gradients in consolidated lung regions compared to non-consolidated regions. These results indicate that spatial variability in shear stiffness alters local strain and strain gradient magnitudes in people with Cystic Fibrosis. This imaged-based modeling technique therefore represents a clinically viable way to non-invasively assess lung mechanics during both health and disease.

19.
Lab Chip ; 24(18): 4390-4402, 2024 09 10.
Article in English | MEDLINE | ID: mdl-39161999

ABSTRACT

Patients with compromised respiratory function frequently require mechanical ventilation to survive. Unfortunately, non-uniform ventilation of injured lungs generates complex mechanical forces that lead to ventilator induced lung injury (VILI). Although investigators have developed lung-on-a-chip systems to simulate normal respiration, modeling the complex mechanics of VILI as well as the subsequent recovery phase is a challenge. Here we present a novel humanized in vitro ventilator-on-a-chip (VOC) model of the lung microenvironment that simulates the different types of injurious forces generated in the lung during mechanical ventilation. We used transepithelial/endothelial electrical impedance measurements to investigate how individual and simultaneous application of mechanical forces alters real-time changes in barrier integrity during and after injury. We find that compressive stress (i.e. barotrauma) does not significantly alter barrier integrity while over-distention (20% cyclic radial strain, volutrauma) results in decreased barrier integrity that quickly recovers upon removal of mechanical stress. Conversely, surface tension forces generated during airway reopening (atelectrauma), result in a rapid loss of barrier integrity with a delayed recovery relative to volutrauma. Simultaneous application of cyclic stretching (volutrauma) and airway reopening (atelectrauma), indicates that the surface tension forces associated with reopening fluid-occluded lung regions are the primary driver of barrier disruption. Thus, our novel VOC system can monitor the effects of different types of injurious forces on barrier disruption and recovery in real-time and can be used to interogate the biomechanical mechanisms of VILI.


Subject(s)
Lab-On-A-Chip Devices , Respiration, Artificial , Ventilator-Induced Lung Injury , Humans , Ventilator-Induced Lung Injury/etiology , Respiration, Artificial/instrumentation , Respiration, Artificial/adverse effects , Electric Impedance , Stress, Mechanical , Lung
20.
bioRxiv ; 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38464068

ABSTRACT

Patients with compromised respiratory function frequently require mechanical ventilation to survive. Unfortunately, non-uniform ventilation of injured lungs generates complex mechanical forces that lead to ventilator induced lung injury (VILI). Although investigators have developed lung-on-a-chip systems to simulate normal respiration, modeling the complex mechanics of VILI as well as the subsequent recovery phase is a challenge. Here we present a novel humanized in vitro ventilator-on-a-chip (VOC) model of the lung microenvironment that simulates the different types of injurious forces generated in the lung during mechanical ventilation. We used transepithelial/endothelial electrical resistance (TEER) measurements to investigate how individual and simultaneous application of the different mechanical forces alters real-time changes in barrier integrity during and after injury. We find that compressive stress (i.e. barotrauma) does not significantly alter barrier integrity while over-distention (20% cyclic radial strain, volutrauma) results in decreased barrier integrity that quickly recovers upon removal of mechanical stress. Conversely, surface tension forces generated during airway reopening (atelectrauma), result in a rapid loss of barrier integrity with a delayed recovery relative to volutrauma. Simultaneous application of cyclic stretching (volutrauma) and airway reopening (atelectrauma), indicate that the surface tension forces associated with reopening fluid-occluded lung regions is the primary driver of barrier disruption. Thus, our novel VOC system can monitor the effects of different types of injurious forces on barrier disruption and recovery in real-time and can be used to identify the biomechanical mechanisms of VILI.

SELECTION OF CITATIONS
SEARCH DETAIL