Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 489
Filter
1.
Allergol Immunopathol (Madr) ; 52(4): 53-59, 2024.
Article in English | MEDLINE | ID: mdl-38970265

ABSTRACT

BACKGROUND: Pulmonary fibrosis is a pathological hallmark of lung injury. It is an aggressive disease that replaces normal lung parenchyma by fibrotic tissue. The transforming growth factor-beta-mothers against decapentaplegic homolog 3 (TGF-ß1-Smad3) signaling pathway plays a key role in regulating lung fibrosis. Decorin (DCN), a small leucine-rich proteoglycan, has a modulatory effect on the immune system by reversibly binding with TGF-ß and reducing its bioavailability. Mesenchymal stem cell (MSC) therapy is a new strategy that has an immune-modulatory capacity. OBJECTIVE: The aim of this study was to introduce a new therapeutic approach to harness remodeling in injured lung. MATERIAL AND METHODS: Bone marrow MSCs were isolated and transduced by decorin gene. Lung injury was induced by bleomycin and mice were treated with MSCs, MSCs-decorin, and decorin. Then, oxidative stress biomarkers, remodeling biomarkers, bronchoalveolar lavage cells, and histopathology study were conducted. RESULTS: Reduced catalase and superoxide dismutase increased due to treatments. Elevated malondialdehyde, hydroxyproline, TGF-ß levels, and polymorphonuclear cells count decreased in the treated groups. Additionally, the histopathology of lung tissues showed controlled inflammation and fibrosis. CONCLUSION: Transfected decorin gene to MSCs and used cell therapy could control remodeling and bleomycin-induced lung injury.


Subject(s)
Bleomycin , Decorin , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Pulmonary Fibrosis , Decorin/genetics , Decorin/metabolism , Animals , Mice , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/therapy , Lung Injury/chemically induced , Lung Injury/therapy , Lung Injury/immunology , Lung Injury/genetics , Transduction, Genetic , Oxidative Stress , Cells, Cultured , Disease Models, Animal , Male , Humans
2.
Stem Cells Transl Med ; 13(7): 606-624, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38819251

ABSTRACT

INTRODUCTION: Lung injuries, such as bronchopulmonary dysplasia (BPD), remain a major complication of preterm birth, with limited therapeutic options. One potential emerging therapy is umbilical cord blood (UCB)-derived therapy. OBJECTIVES: To systematically assess the safety and efficacy of UCB-derived therapy for preterm lung injury in preclinical and clinical studies. METHODS: A systematic search of MEDLINE, Embase, CENTRAL, ClinicalTrials.gov, and WHO International Trials Registry Platform was performed. A meta-analysis was conducted with Review Manager (5.4.1) using a random effects model. Data was expressed as standardized mean difference (SMD) for preclinical data and pooled relative risk (RR) for clinical data, with 95% confidence intervals (CI). Potential effect modifiers were investigated via subgroup analysis. Certainty of evidence was assessed using the GRADE system. RESULTS: Twenty-three preclinical studies and six clinical studies met eligibility criteria. Statistically significant improvements were seen across several preclinical outcomes, including alveolarization (SMD, 1.32, 95%CI [0.99, 1.65]), angiogenesis (SMD, 1.53, 95%CI [0.87, 2.18]), and anti-inflammatory cytokines (SMD, 1.68, 95%CI [1.03, 2.34]). In clinical studies, 103 preterm infants have received UCB-derived therapy for preterm lung injury and no significant difference was observed in the development of BPD (RR, 0.93, 95%CI [0.73, 1.18]). Across both preclinical and clinical studies, administration of UCB-derived therapy appeared safe. Certainty of evidence was assessed as "low." CONCLUSIONS: Administration of UCB-derived therapy was associated with statistically significant improvements across several lung injury markers in preclinical studies. Early clinical studies demonstrated the administration of UCB-derived therapy as safe and feasible but lacked data regarding efficacy.


Subject(s)
Fetal Blood , Humans , Fetal Blood/cytology , Bronchopulmonary Dysplasia/therapy , Infant, Newborn , Infant, Premature , Lung Injury/therapy , Animals , Cord Blood Stem Cell Transplantation/methods
4.
ACS Nano ; 18(20): 13361-13376, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38728619

ABSTRACT

Oxygen therapy cannot rescue local lung hypoxia in patients with severe respiratory failure. Here, an inhalable platform is reported for overcoming the aberrant hypoxia-induced immune changes and alveolar damage using camouflaged poly(lactic-co-glycolic) acid (PLGA) microparticles with macrophage apoptotic body membrane (cMAB). cMABs are preloaded with mitochondria-targeting superoxide dismutase/catalase nanocomplexes (NCs) and modified with pathology-responsive macrophage growth factor colony-stimulating factor (CSF) chains, which form a core-shell platform called C-cMAB/NC with efficient deposition in deeper alveoli and high affinity to alveolar epithelial cells (AECs) after CSF chains are cleaved by matrix metalloproteinase 9. Therefore, NCs can be effectively transported into mitochondria to inhibit inflammasome-mediated AECs damage in mouse models of hypoxic acute lung injury. Additionally, the at-site CSF release is sufficient to rescue circulating monocytes and macrophages and alter their phenotypes, maximizing synergetic effects of NCs on creating a pro-regenerative microenvironment that enables resolution of lung injury and inflammation. This inhalable platform may have applications to numerous inflammatory lung diseases.


Subject(s)
Macrophages , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Mice , Macrophages/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Mice, Inbred C57BL , Hypoxia , Acute Lung Injury/pathology , Lung Injury/pathology , Lung Injury/therapy , Administration, Inhalation , Apoptosis/drug effects
5.
Stem Cell Res Ther ; 15(1): 147, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38773627

ABSTRACT

BACKGROUND: Bleomycin (BLM)-induced lung injury is characterized by mixed histopathologic changes with inflammation and fibrosis, such as observed in human patients with bronchopulmonary dysplasia, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. Although no curative therapies for these lung diseases exist, stem cell therapy has emerged as a potential therapeutic option. Multilineage-differentiating stress-enduring (Muse) cells are endogenous pluripotent- and macrophage-like stem cells distributed in various adult and fetal tissues as stage-specific embryonic antigen-3-positive cells. They selectively home to damaged tissue by sensing sphingosine-1-phosphate and replace the damaged/apoptotic cells by in vivo differentiation. Clinical trials for some human diseases suggest the safety and therapeutic efficacy of intravenously injected human leukocyte antigen-mismatched allogenic Muse cells from adult bone marrow (BM) without immunosuppressant. Here, we evaluated the therapeutic effects of human Muse cells from preterm and term umbilical cord (UC), and adult BM in a rat BLM-induced lung injury model. METHODS: Rats were endotracheally administered BLM to induce lung injury on day 0. On day 3, human preterm UC-Muse, term UC-Muse, or adult BM-Muse cells were administered intravenously without immunosuppressants, and rats were subjected to histopathologic analysis on day 21. Body weight, serum surfactant protein D (SP-D) levels, and oxygen saturation (SpO2) were monitored. Histopathologic lung injury scoring by the Ashcroft and modified American Thoracic Society document scales, quantitative characterization of engrafted Muse cells, RNA sequencing analysis, and in vitro migration assay of infused Muse cells were performed. RESULTS: Rats administered preterm- and term-UC-Muse cells exhibited a significantly better recovery based on weight loss, serum SP-D levels, SpO2, and histopathologic lung injury scores, and a significantly higher rate of both Muse cell homing to the lung and alveolar marker expression (podoplanin and prosurfactant protein-C) than rats administered BM-Muse cells. Rats receiving preterm-UC-Muse cells showed statistically superior results to those receiving term-UC-Muse cells in many of the measures. These findings are thought to be due to higher expression of genes related to cell migration, lung differentiation, and cell adhesion. CONCLUSION: Preterm UC-Muse cells deliver more efficient therapeutic effects than term UC- and BM-Muse cells for treating BLM-induced lung injury in a rat model.


Subject(s)
Bleomycin , Disease Models, Animal , Lung Injury , Umbilical Cord , Animals , Humans , Rats , Lung Injury/therapy , Lung Injury/chemically induced , Lung Injury/pathology , Umbilical Cord/cytology , Rats, Sprague-Dawley , Male , Cell Differentiation , Female
6.
Int J Biochem Cell Biol ; 170: 106568, 2024 May.
Article in English | MEDLINE | ID: mdl-38518980

ABSTRACT

Regulatory T cells, characterized by their expression of the transcription factor Forkhead box P3, are indispensable in maintaining immune homeostasis. The respiratory system is constantly exposed to many environmental challenges, making it susceptible to various insults and infections. Regulatory T cells play essential roles in maintaining homeostasis in the lung and promoting repair after injury. Regulatory T cell function dysregulation can lead to inflammation, tissue damage, or aberrant repair. Research on regulatory T cell mechanisms in the lung has unveiled their influence on lung inflammation and repair mechanisms. In this review, our goal is to highlight the advances in regulatory T cell biology with respect to lung injury and resolution. We further provide a perspective that a deeper understanding of regulatory T cell interactions in the lung microenvironment in health and disease states offers opportunities for therapeutic interventions as treatments to promote lung health.


Subject(s)
Lung Injury , Humans , Lung Injury/therapy , T-Lymphocytes, Regulatory , Lung/metabolism , Inflammation/metabolism , Homeostasis , Forkhead Transcription Factors/metabolism
7.
Stem Cell Res Ther ; 15(1): 80, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38486338

ABSTRACT

BACKGROUND: Despite advances in neonatal care, the incidence of Bronchopulmonary Dysplasia (BPD) remains high among preterm infants. Human induced pluripotent stem cells (hiPSCs) have shown promise in repairing injury in animal BPD models. Evidence suggests they exert their effects via paracrine mechanisms. We aim herein to assess the effectiveness of extracellular vesicles (EVs) derived from hiPSCs and their alveolar progenies (diPSCs) in attenuating hyperoxic injury in a preterm lung explant model. METHODS: Murine lung lobes were harvested on embryonic day 17.5 and maintained in air-liquid interface. Following exposure to 95% O2 for 24 h, media was supplemented with 5 × 106 particles/mL of EVs isolated from hiPSCs or diPSCs by size-exclusion chromatography. On day 3, explants were assessed using Hematoxylin-Eosin staining with mean linear intercept (MLI) measurements, immunohistochemistry, VEGFa and antioxidant gene expression. Statistical analysis was conducted using one-way ANOVA and Multiple Comparison Test. EV proteomic profiling was performed, and annotations focused on alveolarization and angiogenesis signaling pathways, as well as anti-inflammatory, anti-oxidant, and regenerative pathways. RESULTS: Exposure of fetal lung explants to hyperoxia induced airspace enlargement, increased MLI, upregulation of anti-oxidants Prdx5 and Nfe2l2 with decreased VEGFa expression. Treatment with hiPSC-EVs improved parenchymal histologic changes. No overt changes in vasculature structure were observed on immunohistochemistry in our in vitro model. However, VEGFa and anti-oxidant genes were upregulated with diPSC-EVs, suggesting a pro-angiogenic and cytoprotective potential. EV proteomic analysis provided new insights in regard to potential pathways influencing lung regeneration. CONCLUSION: This proof-of-concept in vitro study reveals a potential role for hiPSC- and diPSC-EVs in attenuating lung changes associated with prematurity and oxygen exposure. Our findings pave the way for a novel cell free approach to prevent and/or treat BPD, and ultimately reduce the global burden of the disease.


Subject(s)
Bronchopulmonary Dysplasia , Extracellular Vesicles , Hyperoxia , Induced Pluripotent Stem Cells , Lung Injury , Animals , Mice , Humans , Infant, Newborn , Hyperoxia/complications , Hyperoxia/metabolism , Hyperoxia/pathology , Animals, Newborn , Induced Pluripotent Stem Cells/metabolism , Lung Injury/therapy , Lung Injury/etiology , Antioxidants/metabolism , Proteomics , Infant, Premature , Lung/pathology , Bronchopulmonary Dysplasia/therapy , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Extracellular Vesicles/metabolism
8.
Arch Toxicol ; 98(5): 1297-1310, 2024 May.
Article in English | MEDLINE | ID: mdl-38498160

ABSTRACT

Lung injury has been a serious medical problem that requires new therapeutic approaches and biomarkers. Circular RNAs (circRNAs) are non-coding RNAs (ncRNAs) that exist widely in eukaryotes. CircRNAs are single-stranded RNAs that form covalently closed loops. CircRNAs are significant gene regulators that have a role in the development, progression, and therapy of lung injury by controlling transcription, translating into protein, and sponging microRNAs (miRNAs) and proteins. Although the study of circRNAs in lung injury caused by pulmonary toxicants is just beginning, several studies have revealed their expression patterns. The function that circRNAs perform in relation to pulmonary toxicants (severe acute respiratory distress syndrome coronavirus-2 (SARS-CoV-2), drug abuse, PM2.5, and cigarette smoke) is the main topic of this review. A variety of circRNAs can serve as potential biomarkers of lung injury. In this review, the biogenesis, properties, and biological functions of circRNAs were concluded, and the relationship between circRNAs and pulmonary toxicants was discussed. It is expected that the new ideas and potential treatment targets that circRNAs provide would be beneficial to research into the molecular mechanisms behind lung injury.


Subject(s)
Lung Injury , MicroRNAs , Humans , RNA, Circular/genetics , Lung Injury/chemically induced , Lung Injury/genetics , Lung Injury/therapy , Lung/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Biomarkers/metabolism
9.
Int Immunopharmacol ; 130: 111519, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38442573

ABSTRACT

This study investigates the molecular mechanisms by which extracellular vesicles (EVs) derived from adipose-derived mesenchymal stem cells (ADSCs) promote M2 polarization of macrophages and thus reduce lung injury caused by sepsis. High-throughput sequencing was used to identify differentially expressed genes related to long non-coding RNA (lncRNA) in ADSC-derived EVs (ADSC-EVs) in sepsis lung tissue. Weighted gene co-expression network analysis (WGCNA) was employed to predict the downstream target genes of the lncRNA DLEU2. The RNAInter database predicted miRNAs that interact with DLEU2 and LXN. Functional and pathway enrichment analyses were performed using GO and KEGG analysis. A mouse model of sepsis was established, and treatment with a placebo or ADSC-EVs was administered, followed by RT-qPCR analysis. ADSC-EVs were isolated and identified. In vitro cell experiments were conducted using the mouse lung epithelial cell line MLE-12, mouse macrophage cell line RAW264.7, and mouse lung epithelial cell line (LEPC). ADSC-EVs were co-cultured with RAW264.7 and MLE-12/LEPC cells to study the regulatory mechanism of the lncRNA DLEU2. Cell viability, proliferation, and apoptosis of lung injury cells were assessed using CCK-8, EdU, and flow cytometry. ELISA was used to measure the levels of inflammatory cytokines in the sepsis mouse model, flow cytometry was performed to determine the number of M1 and M2 macrophages, lung tissue pathology was evaluated by H&E staining, and immunohistochemistry was conducted to examine the expression of proliferation- and apoptosis-related proteins. High-throughput sequencing and bioinformatics analysis revealed enrichment of the lncRNA DLEU2 in ADSC-EVs in sepsis lung tissue. Animal and in vitro cell experiments showed increased expression of the lncRNA DLEU2 in sepsis lung tissue after treatment with ADSC-EVs. Furthermore, ADSC-EVs were found to transfer the lncRNA DLEU2 to macrophages, promoting M2 polarization, reducing inflammation response in lung injury cells, and enhancing their viability, proliferation, and apoptosis inhibition. Further functional experiments indicated that lncRNA DLEU2 promotes M2 polarization of macrophages by regulating miR-106a-5p/LXN, thereby enhancing the viability and proliferation of lung injury cells and inhibiting apoptosis. Overexpression of miR-106a-5p could reverse the biological effects of ADSC-EVs-DLEU2 on MLE-12 and LEPC in vitro cell models. Lastly, in vivo animal experiments confirmed that ADSC-EVs-DLEU2 promotes high expression of LXN by inhibiting the expression of miR-106a-5p, further facilitating M2 macrophage polarization and reducing lung edema, thus alleviating sepsis-induced lung injury. lncRNA DLEU2 in ADSC-EVs may promote M2 polarization of macrophages and enhance the viability and proliferation of lung injury cells while inhibiting inflammation and apoptosis reactions, thus ameliorating sepsis-induced lung injury in a mechanism involving the regulation of the miR-106a-5p/LXN axis.


Subject(s)
Lung Injury , MicroRNAs , Nerve Tissue Proteins , RNA, Long Noncoding , Sepsis , Animals , Mice , Apoptosis/genetics , Disease Models, Animal , Lung Injury/microbiology , Lung Injury/therapy , MicroRNAs/genetics , RNA, Long Noncoding/administration & dosage , RNA, Long Noncoding/genetics , Sepsis/complications , Sepsis/genetics , Nerve Tissue Proteins/genetics , Mesenchymal Stem Cells , Exosomes , Male , Mice, Inbred C57BL
11.
Mol Med Rep ; 29(1)2024 01.
Article in English | MEDLINE | ID: mdl-37997800

ABSTRACT

Perinatal exposure of the neonatal lung to inflammation leads to decreased lung angiogenesis and the development of bronchopulmonary dysplasia (BPD). Notably, autologous cord blood mononuclear cells (ACBMNCs) can substantially prevent severe BPD and decrease the inflammatory response in surviving very preterm neonates. Angiopoietin­like protein 7 (Angptl7) is one of the main paracrine cytokines in cord blood stem cells, and is capable of stimulating human hematopoietic stem and progenitor cell expansion. The present study compared Angptl7 levels between the ACBMNCs infusion and control groups (cohort 1). Subsequently, the association between cord blood Angptl7 levels and BPD incidence in a cohort of very preterm neonates was assessed (cohort 2). The hypothesis was further verified in a lipopolysaccharide (LPS)­induced lung injury mouse model. The mRNA expression levels and protein concentrations of inflammatory cytokines in the lung tissue and mouse serum were measured using reverse transcription­quantitative PCR and ELISA, respectively. The number and diameter of lung vessels and macrophage infiltration were assessed using immunofluorescence staining. Compared with in the control group, Angptl7 levels were significantly higher in the ACBMNCs infusion group in cohort 1. In cohort 2, the cord blood Angptl7 levels were significantly lower in infants who later developed BPD. Multiple linear regression analysis showed that higher Angptl7 level was an independent protective factor for BPD. The concentrations of interleukin­6 and monocyte chemoattractant protein­1 were negatively correlated with cord blood Angptl7 level; whereas, vascular endothelial growth factor­A levels were positively correlated with Angptl7 levels. In the LPS­induced lung injury mouse model, the LPS group presented with a significant loss of pulmonary vessels and smaller vessel diameters, which were ameliorated in the Angptl7 treatment group. Furthermore, LPS­induced lung inflammation and macrophage infiltration were alleviated by Angptl7 treatment (P<0.05). In conclusion, the anti­inflammatory and proangiogenic effects of Angptl7 derived from cord blood stem cells may ameliorate BPD severity. The trial for cohort 1 was registered at ClinicalTrials.gov (trial registration no. NCT02999373; date registered, December 21, 2016).


Subject(s)
Bronchopulmonary Dysplasia , Lung Injury , Infant, Newborn , Infant , Pregnancy , Female , Humans , Animals , Mice , Bronchopulmonary Dysplasia/genetics , Vascular Endothelial Growth Factor A , Angiopoietin-Like Protein 7/genetics , Lung Injury/therapy , Lung Injury/complications , Fetal Blood , Lipopolysaccharides , Stem Cells , Cytokines , Anti-Inflammatory Agents
12.
Cells ; 12(23)2023 11 29.
Article in English | MEDLINE | ID: mdl-38067158

ABSTRACT

This study investigates the therapeutic potential of human placental mesenchymal stem cells (P-MSCs) and their extracellular vesicles (EVs) in a murine model of acute respiratory distress syndrome (ARDS), a condition with growing relevance due to its association with severe COVID-19. We induced ARDS-like lung injury in mice using intranasal LPS instillation and evaluated histological changes, neutrophil accumulation via immunohistochemistry, bronchoalveolar lavage fluid cell count, total protein, and cytokine concentration, as well as lung gene expression changes at three time points: 24, 72, and 168 h. We found that both P-MSCs and EV treatments reduced the histological evidence of lung injury, decreased neutrophil infiltration, and improved alveolar barrier integrity. Analyses of cytokines and gene expression revealed that both treatments accelerated inflammation resolution in lung tissue. Biodistribution studies indicated negligible cell engraftment, suggesting that intraperitoneal P-MSC therapy functions mostly through soluble factors. Overall, both P-MSC and EV therapy ameliorated LPS-induced lung injury. Notably, at the tested dose, EV therapy was more effective than P-MSCs in reducing most aspects of lung injury.


Subject(s)
Extracellular Vesicles , Lung Injury , Mesenchymal Stem Cells , Respiratory Distress Syndrome , Pregnancy , Humans , Animals , Female , Mice , Lung Injury/therapy , Disease Models, Animal , Lipopolysaccharides/metabolism , Tissue Distribution , Placenta/metabolism , Respiratory Distress Syndrome/therapy , Respiratory Distress Syndrome/metabolism , Extracellular Vesicles/metabolism , Cytokines/metabolism , Mesenchymal Stem Cells/metabolism
13.
Ter Arkh ; 95(7): 591-596, 2023 Sep 29.
Article in Russian | MEDLINE | ID: mdl-38159011

ABSTRACT

Vaping, i.e. the use of electronic nicotine/other substances delivery systems, increases a risk of vaping-associated lung injury. The review describes clinical manifestation, methods of diagnosis and diagnostic criteria, treatment of patients with this disease as well as risk stratification of vapers and approaches to their management based on Worchester classification and clinical guidance. The pathogenetic mechanisms of vaping-associated lung injury have been analyzed.


Subject(s)
Electronic Nicotine Delivery Systems , Lung Injury , Vaping , Humans , Lung Injury/diagnosis , Lung Injury/etiology , Lung Injury/therapy , Vaping/adverse effects , Tomography, X-Ray Computed
14.
Cell Rep ; 42(7): 112745, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37405911

ABSTRACT

Although increasing evidence suggests potential iatrogenic injury from supplemental oxygen therapy, significant exposure to hyperoxia in critically ill patients is inevitable. This study shows that hyperoxia causes lung injury in a time- and dose-dependent manner. In addition, prolonged inspiration of oxygen at concentrations higher than 80% is found to cause redox imbalance and impair alveolar microvascular structure. Knockout of C-X-C motif chemokine receptor 1 (Cxcr1) inhibits the release of reactive oxygen species (ROS) from neutrophils and synergistically enhances the ability of endothelial cells to eliminate ROS. We also combine transcriptome, proteome, and metabolome analysis and find that CXCR1 knockdown promotes glutamine metabolism and leads to reduced glutathione by upregulating the expression of malic enzyme 1. This preclinical evidence suggests that a conservative oxygen strategy should be recommended and indicates that targeting CXCR1 has the potential to restore redox homeostasis by reducing oxygen toxicity when inspiratory hyperoxia treatment is necessary.


Subject(s)
Hyperoxia , Lung Injury , Receptors, Interleukin-8A , Humans , Endothelial Cells/metabolism , Glutamine/metabolism , Hyperoxia/complications , Hyperoxia/metabolism , Lung/metabolism , Lung Injury/therapy , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Animals , Mice , Receptors, Interleukin-8A/metabolism
15.
J Am Board Fam Med ; 36(3): 405-413, 2023 May 08.
Article in English | MEDLINE | ID: mdl-37290827

ABSTRACT

BACKGROUND: Recently, the use of electronic cigarettes increased sharply, leading to increased e-cigarette, or Vaping Product Use-Associated Lung Injury (EVALI), and other acute pulmonary conditions. There is an urgent need for clinical information about e-cigarette users to identify factors that contribute to EVALI. We developed an e-cigarette/vaping assessment tool (EVAT) that was integrated into the Electronic Health Record (EHR) of a large state-wide medical system and initiated a system-wide dissemination and education to support its use. METHODS: EVAT documented current vaping status, history, and e-cigarette content (nicotine, cannabinoids, and/or flavoring). Educational materials and presentations were developed via a comprehensive literature review. EVAT utilization in the EHR was assessed quarterly. Patients' demographic data and clinical site name were also collected. RESULTS: The EVAT was built, validated, and integrated with the EHR in July 2020. Live and virtual seminars were conducted for prescribing providers and clinical staff. Asynchronous training was offered using podcasts, e-mails, and Epic tip sheets. Participants were informed about vaping harm and EVALI and instructed on the use of EVAT. As of December 31, 2022, EVAT was used 988,181 times, with 376,559 unique patients evaluated. Overall, 1,063 hospital units and affiliated ambulatory clinics used EVAT, including 64 Primary Care, 95 Pediatrics, and 874 Specialty sites. CONCLUSIONS: EVAT was successfully implemented. Continued outreach efforts are needed to further increase its usage. Education materials should be enhanced to help providers to reach youth and vulnerable populations and connect patients to the tobacco treatment resources.


Subject(s)
Electronic Nicotine Delivery Systems , Lung Injury , Vaping , Adolescent , Humans , Child , Lung Injury/therapy , Vaping/adverse effects , Electronic Health Records , Nicotine
16.
Immunol Invest ; 52(6): 681-702, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37310728

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia (BPD) is the predominant chronic disorder in preterm neonates. This study explored impacts of miR-34c-5p carried by bone marrow stromal cells-secreted extracellular vesicles (BMSC-EVs) on BPD progression. METHODS: A BPD mouse model was established, followed by measurement of miR-34c-5p, OTUD3, and PTEN expression. EVs were isolated from BMSCs transfected with miR-34c-5p mimic or mimic NC and intratracheally injected into mice. CD31 and Ki67 expression was detected and the pathological changes of lung tissues and lung function indexes were observed for mice. A neonatal human pulmonary microvascular endothelial cell (HPMEC) model was developed with hyperoxia, followed by co-culture with extracted EVs and ectopic experiments for measurement of cell viability, migration, and angiogenesis. IL-4, IL-13, IL-1ß, and IL-6 levels were measured in cell supernatants and lung tissues. Dual-luciferase reporter, ubiquitination, Co-IP, and RIP assays were adopted to determine the relationship among miR-34c-5p, OTUD3, and PTEN. RESULTS: Lung tissues of BPD mice had downregulated miR-34c-5p expression and upregulated OTUD3 and PTEN expression. BMSC-EVs and BMSC-EVs-miR-34c-5p treatment improved lung injury and alveolar structure, decreased lung resistance and IL-4, IL-13, IL-1ß, and IL-6 levels, and elevated dynamic lung compliance in BPD mice, as well as enhanced proliferation, angiogenesis, and migration and restrained inflammation in HPMECs. Mechanistically, miR-34c-5p negatively targeted OTUD3 which restrained ubiquitination to promote PTEN protein stabilization. Upregulation of OTUD3 or PTEN negated the changes in the proliferation, angiogenesis, migration, and inflammation of hyperoxia-treated HPMECs induced by BMSC-EVs-miR-34c-5p. CONCLUSION: BMSC-EVs-miR-34c-5p alleviated lung injury and inflammation in hyperoxia-induced BPD by blocking the OTUD3/PTEN axis.


Subject(s)
Bronchopulmonary Dysplasia , Extracellular Vesicles , Hyperoxia , Lung Injury , Mesenchymal Stem Cells , MicroRNAs , Infant, Newborn , Humans , Animals , Mice , Bronchopulmonary Dysplasia/therapy , Bronchopulmonary Dysplasia/metabolism , Lung Injury/therapy , Lung Injury/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Interleukin-13/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Hyperoxia/metabolism , Interleukin-4 , Interleukin-6/metabolism , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Inflammation/metabolism , Ubiquitin-Specific Proteases/metabolism
17.
Tumori ; 109(6): NP11-NP13, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37165581

ABSTRACT

Electronic cigarette, or vaping, product use-associated lung injury (EVALI) is an increasingly recognized entity with the potential for severe pulmonary toxicity. We present the case of a young man first evaluated at a tertiary care center in the United States in 2019 with newly diagnosed testicular cancer with acute respiratory failure, which was initially attributed to possible metastatic disease but eventually determined to be related to EVALI. This case highlights the clinical features of EVALI, the potential diagnostic dilemma that can arise with EVALI when occurring in the setting of malignancy and the importance of inquiring about vaping use among patients with malignancy, especially in adolescents and young adults.


Subject(s)
Electronic Nicotine Delivery Systems , Lung Injury , Neoplasms, Germ Cell and Embryonal , Testicular Neoplasms , Vaping , Male , Adolescent , Young Adult , Humans , United States , Lung Injury/diagnosis , Lung Injury/etiology , Lung Injury/therapy , Testicular Neoplasms/diagnosis , Testicular Neoplasms/etiology , Vaping/adverse effects , Neoplasms, Germ Cell and Embryonal/complications
18.
Indian J Pediatr ; 90(5): 470-480, 2023 05.
Article in English | MEDLINE | ID: mdl-37010692

ABSTRACT

Providing the right respiratory support is an essential skill, vital for anyone treating sick children. Recent advances in respiratory support include developments in both non-invasive and invasive ventilatory strategies. In non-invasive ventilation, newer modalities are being developed, in an attempt to decrease the need for invasive ventilation. This include newer techniques like Heated humidified high-flow nasal cannula (HHHFNC) and improvements in existing modes. The success of Continuous positive airway pressure (CPAP) and other non-invasive modes depend to a large extent on choosing and maintaining a suitable interface. When it comes to invasive ventilation, recent advances are focussing on increasing automation, improving patient comfort and minimising lung injury. Concepts like mechanical power are attempts at understanding the mechanisms of unintended injuries resulting from respiratory support and newer monitoring methods like transpulmonary pressure, thoracic impedance tomography are attempts at measuring potential markers of lung injury. Using the vast arrays of available ventilatory options judiciously, considering their advantages and drawbacks in every individual case will be the prime responsibility of clinicians in the future. Simultaneously, efforts have been made to identify potential drugs that can favourably modify the pathophysiology of acute respiratory distress syndrome (ARDS). Unfortunately, though eagerly awaited, most pharmaceutical agents tried in pediatric ARDS have not shown definite benefit. Pulmonary local drug and gene therapy using liquid ventilation strategies may revolutionize our future understanding and management of lung diseases.


Subject(s)
Lung Injury , Respiratory Distress Syndrome, Newborn , Respiratory Distress Syndrome , Respiratory Insufficiency , Infant, Newborn , Humans , Child , Respiratory Distress Syndrome, Newborn/therapy , Lung Injury/therapy , Respiration, Artificial , Continuous Positive Airway Pressure/methods , Respiratory Insufficiency/therapy , Respiratory Distress Syndrome/therapy , Oxygen Inhalation Therapy/methods
19.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 35(2): 217-220, 2023 Feb.
Article in Chinese | MEDLINE | ID: mdl-36916386

ABSTRACT

Mechanical ventilation is an advanced life support treatment for patients with acute respiratory failure. While stabilizing respiratory function, it also acts as an injury factor to exacerbate or lead to lung injury, that is, ventilation-induced lung injury (VILI). There may be a more subtle form of damage to VILI known as "biotrauma". However, the mechanism of biotrauma in VILI is still unclear. This article intends to review the mechanism of biotrauma of VILI from the aspects of inflammatory response, oxidative stress and complement activation, in order to provide a new strategy for clinical prevention and treatment of biotrauma caused by VILI.


Subject(s)
Lung Injury , Respiratory Distress Syndrome , Ventilator-Induced Lung Injury , Humans , Lung Injury/etiology , Lung Injury/therapy , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/prevention & control , Lung , Respiratory Distress Syndrome/therapy , Respiratory Physiological Phenomena
SELECTION OF CITATIONS
SEARCH DETAIL
...