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.
PLoS Biol ; 22(4): e3002566, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38652717

ABSTRACT

Phage therapy is a therapeutic approach to treat multidrug-resistant (MDR) infections that employs lytic bacteriophages (phages) to eliminate bacteria. Despite the abundant evidence for its success as an antimicrobial in Eastern Europe, there is scarce data regarding its effects on the human host. Here, we aimed to understand how lytic phages interact with cells of the airway epithelium, the tissue site that is colonized by bacterial biofilms in numerous chronic respiratory disorders. Using a panel of Pseudomonas aeruginosa phages and human airway epithelial cells (AECs) derived from a person with cystic fibrosis (CF), we determined that interactions between phages and epithelial cells depend on specific phage properties as well as physiochemical features of the microenvironment. Although poor at internalizing phages, the airway epithelium responds to phage exposure by changing its transcriptional profile and secreting antiviral and proinflammatory cytokines that correlate with specific phage families. Overall, our findings indicate that mammalian responses to phages are heterogenous and could potentially alter the way that respiratory local defenses aid in bacterial clearance during phage therapy. Thus, besides phage receptor specificity in a particular bacterial isolate, the criteria to select lytic phages for therapy should be expanded to include mammalian cell responses.


Subject(s)
Cystic Fibrosis , Cytokines , Epithelial Cells , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/virology , Epithelial Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/immunology , Cytokines/metabolism , Cystic Fibrosis/therapy , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Phage Therapy , Bacteriophages/physiology , Bacteriophages/genetics , Respiratory Mucosa/virology , Respiratory Mucosa/metabolism , Respiratory Mucosa/immunology , Pseudomonas Infections/therapy , Pseudomonas Infections/immunology , Pseudomonas Phages/metabolism , Biofilms
2.
Stem Cells ; 42(3): 230-250, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38183264

ABSTRACT

Chronic inflammation and dysregulated repair mechanisms after epithelial damage have been implicated in chronic obstructive pulmonary disease (COPD). However, the lack of ex vivo-models that accurately reflect multicellular lung tissue hinders our understanding of epithelial-mesenchymal interactions in COPD. Through a combination of transcriptomic and proteomic approaches applied to a sophisticated in vitro iPSC-alveolosphere with fibroblasts model, epithelial-mesenchymal crosstalk was explored in COPD and following SARS-CoV-2 infection. These experiments profiled dynamic changes at single-cell level of the SARS-CoV-2-infected alveolar niche that unveiled the complexity of aberrant inflammatory responses, mitochondrial dysfunction, and cell death in COPD, which provides deeper insights into the accentuated tissue damage/inflammation/remodeling observed in patients with SARS-CoV-2 infection. Importantly, this 3D system allowed for the evaluation of ACE2-neutralizing antibodies and confirmed the potency of this therapy to prevent SARS-CoV-2 infection in the alveolar niche. Thus, iPSC-alveolosphere cultured with fibroblasts provides a promising model to investigate disease-specific mechanisms and to develop novel therapeutics.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Pulmonary Disease, Chronic Obstructive , Humans , SARS-CoV-2 , Proteomics , Immunotherapy , Inflammation
3.
Lung ; 202(3): 223-232, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38772946

ABSTRACT

We are entering the post-antibiotic era. Antimicrobial resistance (AMR) is a critical problem in chronic lung infections resulting in progressive respiratory failure and increased mortality. In the absence of emerging novel antibiotics to counter AMR infections, bacteriophages (phages), viruses that infect bacteria, have become a promising option for chronic respiratory infections. However, while personalized phage therapy is associated with improved outcomes in individual cases, clinical trials demonstrating treatment efficacy are lacking, limiting the therapeutic potential of this approach for respiratory infections. In this review, we address the current state of phage therapy for managing chronic respiratory diseases. We then discuss how phage therapy may address major microbiologic obstacles which hinder disease resolution of chronic lung infections with current antibiotic-based treatment practices. Finally, we highlight the challenges that must be addressed for successful phage therapy clinical trials. Through this discussion, we hope to expand on the potential of phages as an adjuvant therapy in chronic lung infections, as well as the microbiologic challenges that need to be addressed for phage therapy to expand beyond personalized salvage therapy.


Subject(s)
Phage Therapy , Respiratory Tract Infections , Humans , Phage Therapy/methods , Respiratory Tract Infections/therapy , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Bacteriophages , Chronic Disease , Anti-Bacterial Agents/therapeutic use
4.
Am J Respir Cell Mol Biol ; 68(1): 75-89, 2023 01.
Article in English | MEDLINE | ID: mdl-36125351

ABSTRACT

The mechanisms by which excessive systemic activation of adaptive T lymphocytes, as in cytokine release syndrome (CRS), leads to innate immune cell-mediated acute lung injury (ALI) or acute respiratory distress syndrome, often in the absence of any infection, remains unknown. Here, we investigated the roles of IFN-γ and IL-17A, key T-cell cytokines significantly elevated in patients with CRS, in the immunopathogenesis of CRS-induced extrapulmonary ALI. CRS was induced in wild-type (WT), IL-17A- and IFN-γ knockout (KO) human leukocyte antigen-DR3 transgenic mice with 10 µg of the superantigen, staphylococcal enterotoxin B, given intraperitoneally. Several ALI parameters, including gene expression profiling in the lungs, were studied 4, 24, or 48 hours later. Systemic T-cell activation with staphylococcal enterotoxin B resulted in robust upregulation of several chemokines, S100A8/A9, matrix metalloproteases, and other molecules implicated in tissue damage, granulocyte as well as agranulocyte adhesion, and diapedesis in the lungs as early as 4 hours, which was accompanied by subsequent neutrophil/eosinophil lung infiltration and severe ALI in IFN-γ KO mice. These pathways were significantly underexpressed in IL-17A KO mice, which manifested mildest ALI and intermediate in WT mice. Neutralization of IFN-γ worsened ALI in WT and IL-17A KO mice, whereas neutralizing IL-17A did not mitigate lung injury in IFN-γ KO mice, suggesting a dominant protective role for IFN-γ in ALI and that IL-17A is dispensable. Ruxolitinib, a Janus kinase inhibitor, increased ALI severity in WT mice. Thus, our study identified novel mechanisms of ALI in CRS and its differential modulation by IFN-γ and IL-17A.


Subject(s)
Acute Lung Injury , Interleukin-17 , Humans , Mice , Animals , Cytokine Release Syndrome , Interferon-gamma , Cytokines , Lung/pathology , Acute Lung Injury/pathology , Mice, Knockout , Mice, Inbred C57BL
5.
Crit Care ; 27(1): 34, 2023 01 23.
Article in English | MEDLINE | ID: mdl-36691080

ABSTRACT

BACKGROUND: Recent single-center reports have suggested that community-acquired bacteremic co-infection in the context of Coronavirus disease 2019 (COVID-19) may be an important driver of mortality; however, these reports have not been validated with a multicenter, demographically diverse, cohort study with data spanning the pandemic. METHODS: In this multicenter, retrospective cohort study, inpatient encounters were assessed for COVID-19 with community-acquired bacteremic co-infection using 48-h post-admission blood cultures and grouped by: (1) confirmed co-infection [recovery of bacterial pathogen], (2) suspected co-infection [negative culture with ≥ 2 antimicrobials administered], and (3) no evidence of co-infection [no culture]. The primary outcomes were in-hospital mortality, ICU admission, and mechanical ventilation. COVID-19 bacterial co-infection risk factors and impact on primary outcomes were determined using multivariate logistic regressions and expressed as adjusted odds ratios with 95% confidence intervals (Cohort, OR 95% CI, Wald test p value). RESULTS: The studied cohorts included 13,781 COVID-19 inpatient encounters from 2020 to 2022 in the University of Alabama at Birmingham (UAB, n = 4075) and Ochsner Louisiana State University Health-Shreveport (OLHS, n = 9706) cohorts with confirmed (2.5%), suspected (46%), or no community-acquired bacterial co-infection (51.5%) and a comparison cohort consisting of 99,170 inpatient encounters from 2010 to 2019 (UAB pre-COVID-19 pandemic cohort). Significantly increased likelihood of COVID-19 bacterial co-infection was observed in patients with elevated ≥ 15 neutrophil-to-lymphocyte ratio (UAB: 1.95 [1.21-3.07]; OLHS: 3.65 [2.66-5.05], p < 0.001 for both) within 48-h of hospital admission. Bacterial co-infection was found to confer the greatest increased risk for in-hospital mortality (UAB: 3.07 [2.42-5.46]; OLHS: 4.05 [2.29-6.97], p < 0.001 for both), ICU admission (UAB: 4.47 [2.87-7.09], OLHS: 2.65 [2.00-3.48], p < 0.001 for both), and mechanical ventilation (UAB: 3.84 [2.21-6.12]; OLHS: 2.75 [1.87-3.92], p < 0.001 for both) across both cohorts, as compared to other risk factors for severe disease. Observed mortality in COVID-19 bacterial co-infection (24%) dramatically exceeds the mortality rate associated with community-acquired bacteremia in pre-COVID-19 pandemic inpatients (5.9%) and was consistent across alpha, delta, and omicron SARS-CoV-2 variants. CONCLUSIONS: Elevated neutrophil-to-lymphocyte ratio is a prognostic indicator of COVID-19 bacterial co-infection within 48-h of admission. Community-acquired bacterial co-infection, as defined by blood culture-positive results, confers greater increased risk of in-hospital mortality, ICU admission, and mechanical ventilation than previously described risk factors (advanced age, select comorbidities, male sex) for COVID-19 mortality, and is independent of SARS-CoV-2 variant.


Subject(s)
Bacteremia , COVID-19 , Coinfection , Community-Acquired Infections , Humans , Male , SARS-CoV-2 , Cohort Studies , Retrospective Studies , Respiration, Artificial , Pandemics , Hospital Mortality , Bacteria , Risk Factors , Intensive Care Units
6.
Am J Physiol Lung Cell Mol Physiol ; 322(6): L822-L841, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35438006

ABSTRACT

Although epithelial-mesenchymal transition (EMT) is a common feature of fibrotic lung disease, its role in fibrogenesis is controversial. Recently, aberrant basaloid cells were identified in fibrotic lung tissue as a novel epithelial cell type displaying a partial EMT phenotype. The developmental origin of these cells remains unknown. To elucidate the role of EMT in the development of aberrant basaloid cells from the bronchial epithelium, we mapped EMT-induced transcriptional changes at the population and single-cell levels. Human bronchial epithelial cells grown as submerged or air-liquid interface (ALI) cultures with or without EMT induction were analyzed by bulk and single-cell RNA-Sequencing. Comparison of submerged and ALI cultures revealed differential expression of 8,247 protein coding (PC) and 1,621 long noncoding RNA (lncRNA) genes and revealed epithelial cell-type-specific lncRNAs. Similarly, EMT induction in ALI cultures resulted in robust transcriptional reprogramming of 6,020 PC and 907 lncRNA genes. Although there was no evidence for fibroblast/myofibroblast conversion following EMT induction, cells displayed a partial EMT gene signature and an aberrant basaloid-like cell phenotype. The substantial transcriptional differences between submerged and ALI cultures highlight that care must be taken when interpreting data from submerged cultures. This work supports that lung epithelial EMT does not generate fibroblasts/myofibroblasts and confirms ALI cultures provide a physiologically relevant system to study aberrant basaloid-like cells and mechanisms of EMT. We provide a catalog of PC and lncRNA genes and an interactive browser (https://bronc-epi-in-vitro.cells.ucsc.edu/) of single-cell RNA-Seq data for further exploration of potential roles in the lung epithelium in health and lung disease.


Subject(s)
Lung Diseases , RNA, Long Noncoding , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelium/metabolism , Humans , Lung Diseases/metabolism , RNA, Long Noncoding/genetics , Respiratory Mucosa/metabolism
7.
Yale J Biol Med ; 95(4): 413-427, 2022 12.
Article in English | MEDLINE | ID: mdl-36568830

ABSTRACT

The rise of antimicrobial resistant (AMR) bacteria is a global public health threat. AMR Achromobacter bacteria pose a challenging clinical problem, particularly for those with cystic fibrosis (CF) who are predisposed to chronic bacterial lung infections. Lytic bacteriophages (phages) offer a potential alternative to treat AMR infections, with the possible benefit that phage selection for resistance in target bacteria might coincide with reduced pathogenicity. The result is a genetic "trade-off," such as increased sensitivity to chemical antibiotics, and/or decreased virulence of surviving bacteria that are phage resistant. Here, we show that two newly discovered lytic phages against Achromobacter were associated with stabilization of respiratory status when deployed to treat a chronic pulmonary infection in a CF patient using inhaled (nebulized) phage therapy. The two phages demonstrate traits that could be generally useful in their development as therapeutics, especially the possibility that the phages can select for clinically useful trade-offs if bacteria evolve phage resistance following therapy. We discuss the limitations of the current study and suggest further work that should explore whether the phages could be generally useful in targeting pulmonary or other Achromobacter infections in CF patients.


Subject(s)
Achromobacter , Bacteriophages , Cystic Fibrosis , Phage Therapy , Humans , Anti-Bacterial Agents/pharmacology , Cystic Fibrosis/therapy , Cystic Fibrosis/complications
8.
Am J Respir Cell Mol Biol ; 61(2): 185-197, 2019 08.
Article in English | MEDLINE | ID: mdl-30742488

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is a versatile human pathogen that is associated with diverse types of infections ranging from benign colonization to sepsis. We postulated that MRSA must undergo specific genotypic and phenotypic changes to cause chronic pulmonary disease. We investigated how MRSA adapts to the human airway to establish chronic infection, as occurs during cystic fibrosis (CF). MRSA isolates from patients with CF that were collected over a 4-year period were analyzed by whole-genome sequencing, transcriptional analysis, and metabolic studies. Persistent MRSA infection was associated with staphylococcal metabolic adaptation, but not changes in immunogenicity. Adaptation was characterized by selective use of the tricarboxylic acid cycle cycle and generation of biofilm, a means of limiting oxidant stress. Increased transcription of specific metabolic genes was conserved in all host-adapted strains, most notably a 10,000-fold increase in fumC, which catalyzes the interconversion of fumarate and malate. Elevated fumarate levels promoted in vitro biofilm production in clinical isolates. Host-adapted strains preferred to assimilate glucose polymers and pyruvate, which can be metabolized to generate N-acetylglucosamine polymers that comprise biofilm. MRSA undergoes substantial metabolic adaptation to the human airway to cause chronic pulmonary infection, and selected metabolites may be useful therapeutically to inhibit infection.


Subject(s)
Cystic Fibrosis/microbiology , Lung Diseases/microbiology , Methicillin-Resistant Staphylococcus aureus/metabolism , Pneumonia, Staphylococcal/microbiology , Staphylococcal Infections/microbiology , Acetylglucosamine/metabolism , Adult , Animals , Biofilms , Bronchi/metabolism , Bronchoalveolar Lavage Fluid , Cystic Fibrosis/metabolism , Cytokines/metabolism , Female , Fumarates/metabolism , Gentamicins/pharmacology , Glucose/metabolism , Humans , Lung Diseases/metabolism , Malates/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phylogeny , Pneumonia, Staphylococcal/metabolism , Pyruvic Acid/metabolism , Staphylococcal Infections/metabolism , Transcription, Genetic , Tricarboxylic Acids/metabolism , Whole Genome Sequencing
9.
J Immunol ; 199(3): 1060-1068, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28646038

ABSTRACT

Influenza viral infections often lead to increased mortality in older people. However, the mechanisms by which aging impacts immunity to influenza lung infection remain unclear. We employed a murine model of influenza infection to identify these mechanisms. With aging, we found reduced numbers of alveolar macrophages, cells essential for lung homeostasis. We also determined that these macrophages are critical for influenza-induced mortality with aging. Furthermore, aging vastly alters the transcriptional profile and specifically downregulates cell cycling pathways in alveolar macrophages. Aging impairs the ability of alveolar macrophages to limit lung damage during influenza infection. Moreover, aging decreases alveolar macrophage phagocytosis of apoptotic neutrophils, downregulates the scavenging receptor CD204, and induces retention of neutrophils during influenza infection. Thus, aging induces defective phagocytosis by alveolar macrophages and increases lung damage. These findings indicate that therapies that enhance the function of alveolar macrophages may improve outcomes in older people infected with respiratory viruses.


Subject(s)
Aging , Influenza, Human/mortality , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Phagocytosis , Animals , Cell Cycle , Disease Models, Animal , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/virology , Lung/immunology , Lung/pathology , Lung/virology , Macrophages, Alveolar/metabolism , Mice , Neutrophils/immunology , Neutrophils/pathology , Neutrophils/virology , Orthomyxoviridae Infections/virology , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism
10.
J Immunol ; 198(7): 2844-2853, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28202616

ABSTRACT

Oxygen is supplied as a supportive treatment for patients suffering from acute respiratory distress syndrome. Unfortunately, high oxygen concentration increases reactive oxygen species generation, which causes DNA damage and ultimately cell death in the lung. Although 8-oxoguanine-DNA glycosylase (OGG-1) is involved in repairing hyperoxia-mediated DNA damage, the underlying molecular mechanism remains elusive. In this study, we report that ogg-1-deficient mice exhibited a significant increase of proinflammatory cytokines (TNF-α, IL-6, and IFN-γ) in the lung after being exposed to 95% oxygen. In addition, we found that ogg-1 deficiency downregulated (macro)autophagy when exposed to hyperoxia both in vitro and in vivo, which was evident by decreased conversion of LC3-I to LC3-II, reduced LC3 punctate staining, and lower Atg7 expression compared with controls. Using a chromatin immunoprecipitation assay, we found that OGG-1 associated with the promoter of Atg7, suggesting a role for OGG1 in regulation of Atg7 activity. Knocking down OGG-1 decreased the luciferase reporter activity of Atg7. Further, inflammatory cytokine levels in murine lung epithelial cell line cells were downregulated following autophagy induction by starvation and rapamycin treatment, and upregulated when autophagy was blocked using 3-methyladenine and chloroquine. atg7 knockout mice and Atg7 small interfering RNA-treated cells exhibited elevated levels of phospho-NF-κB and intensified inflammatory cytokines, suggesting that Atg7 impacts inflammatory responses to hyperoxia. These findings demonstrate that OGG-1 negatively regulates inflammatory cytokine release by coordinating molecular interaction with the autophagic pathway in hyperoxia-induced lung injury.


Subject(s)
Acute Lung Injury/pathology , Autophagy , DNA Glycosylases/metabolism , DNA Repair , Hyperoxia/pathology , Lung/pathology , Acute Lung Injury/metabolism , Animals , Blotting, Western , Chromatin Immunoprecipitation , Comet Assay , Cytokines/biosynthesis , Disease Models, Animal , Hyperoxia/metabolism , Immunoprecipitation , Inflammation/metabolism , Inflammation/pathology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Psychosomatics ; 58(4): 343-354, 2017.
Article in English | MEDLINE | ID: mdl-28576305

ABSTRACT

BACKGROUND: Individuals with cystic fibrosis (CF) are at high risk for depression and anxiety, which are associated with worse medical outcomes. Novel therapies for CF hold great promise for improving physical health, but the effects of these therapies on mental health remain poorly understood. OBJECTIVE: This review aims to familiarize psychiatrists with the potential effect of novel CF therapies on depression and anxiety. METHODS: We discuss novel therapies that directly target the mutant CF protein, the CF transmembrane regulator (CFTR), which are called CFTR modulators. We summarize depression and anxiety screening and treatment guidelines under implementation in accredited CF centers. Case vignettes highlight the complexities of caring for individuals with CF with comorbid depression and anxiety, including patients experiencing worsening depression and anxiety proximate to initiation of CFTR modulator therapy, and management of drug-drug interactions. CONCLUSIONS: Although CFTR modulator therapies provide hope for improving clinical outcomes, worsening depression and anxiety occurs in some patients when starting these novel agents. This phenomenon may be multifactorial, with hypothesized contributions from CFTR modulator-psychotropic medication interactions, direct effects of CFTR modulators on central nervous system function, the psychologic effect of starting a potentially life-altering drug, and typical triggers of depression and anxiety such as stress, pain, and inflammation. The medical and psychiatric complexity of many individuals with CF warrants more direct involvement of mental health specialists on the multidisciplinary CF team. Inclusion of mental health variables in patients with CF registries will facilitate further examination at an epidemiologic level.


Subject(s)
Anxiety Disorders/complications , Chloride Channel Agonists/therapeutic use , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/complications , Cystic Fibrosis/psychology , Depressive Disorder/complications , Anxiety Disorders/drug therapy , Anxiety Disorders/psychology , Attitude to Health , Depressive Disorder/drug therapy , Depressive Disorder/psychology , Humans , Treatment Outcome
12.
Am J Physiol Lung Cell Mol Physiol ; 307(2): L186-96, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24838750

ABSTRACT

Airway epithelial cells are the primary cell type involved in respiratory viral infection. Upon infection, airway epithelium plays a critical role in host defense against viral infection by contributing to innate and adaptive immune responses. Influenza A virus, rhinovirus, and respiratory syncytial virus (RSV) represent a broad range of human viral pathogens that cause viral pneumonia and induce exacerbations of asthma and chronic obstructive pulmonary disease. These respiratory viruses induce airway epithelial production of IL-8, which involves epidermal growth factor receptor (EGFR) activation. EGFR activation involves an integrated signaling pathway that includes NADPH oxidase activation of metalloproteinase, and EGFR proligand release that activates EGFR. Because respiratory viruses have been shown to activate EGFR via this signaling pathway in airway epithelium, we investigated the effect of virus-induced EGFR activation on airway epithelial antiviral responses. CXCL10, a chemokine produced by airway epithelial cells in response to respiratory viral infection, contributes to the recruitment of lymphocytes to target and kill virus-infected cells. While respiratory viruses activate EGFR, the interaction between CXCL10 and EGFR signaling pathways is unclear, and the potential for EGFR signaling to suppress CXCL10 has not been explored. Here, we report that respiratory virus-induced EGFR activation suppresses CXCL10 production. We found that influenza virus-, rhinovirus-, and RSV-induced EGFR activation suppressed IFN regulatory factor (IRF) 1-dependent CXCL10 production. In addition, inhibition of EGFR during viral infection augmented IRF1 and CXCL10. These findings describe a novel mechanism that viruses use to suppress endogenous antiviral defenses, and provide potential targets for future therapies.


Subject(s)
Chemokine CXCL10/biosynthesis , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Interferon Regulatory Factor-1/physiology , Virus Diseases/physiopathology , Bronchi/cytology , Cell Line , Cell Movement/drug effects , ErbB Receptors/antagonists & inhibitors , Female , Gefitinib , Humans , Influenza A Virus, H1N1 Subtype , Interleukin-8/biosynthesis , Killer Cells, Natural/physiology , Quinazolines/pharmacology , Respiratory Syncytial Viruses , Rhinovirus , Signal Transduction
15.
Infect Dis Clin North Am ; 38(1): 149-162, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38280761

ABSTRACT

Patients with cystic fibrosis (CF) often develop respiratory tract infections with pathogenic multidrug-resistant organisms (MDROs) such as methicillin-resistant Staphylococcus aureus, and a variety of gram-negative organisms that include Pseudomonas aeruginosa, Burkholderia sp., Stenotrophomonas maltophilia, Achromobacter xylosoxidans, and nontuberculous mycobacteria (NTM). Despite the introduction of new therapies to address underlying cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction, MDRO infections remain a problem and novel antimicrobial interventions are still needed. Therapeutic approaches include improving the efficacy of existing drugs by adjusting the dose based on differences in CF patient pharmacokinetics/pharmacodynamics, the development of inhaled formulations to reduce systemic adverse events, and the use of newer beta-lactam/beta-lactamase combinations. Alternative innovative therapeutic approaches include the use of gallium and bacteriophages to treat MDRO pulmonary infections including those with extreme antibiotic resistance. However, additional clinical trials are required to determine the optimal dosing and efficacy of these different strategies and to identify patients with CF most likely to benefit from these new treatment options.


Subject(s)
Anti-Infective Agents , Cystic Fibrosis , Methicillin-Resistant Staphylococcus aureus , Respiratory Tract Infections , Stenotrophomonas maltophilia , Humans , Cystic Fibrosis/complications , Cystic Fibrosis/drug therapy , Cystic Fibrosis/microbiology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/microbiology , Anti-Infective Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
16.
J Med Educ Curric Dev ; 11: 23821205241242220, 2024.
Article in English | MEDLINE | ID: mdl-38572090

ABSTRACT

OBJECTIVES: Virtual reality (VR) teaching methods have potential to support medical students acquire increasing amounts of knowledge. EVENT (Easy VR EducatioN Tool) is an open educational resource software for immersive VR environments, which is designed for use without programming skills. In this work, EVENT was used in a medical student VR course on pancreatic cancer. METHODS: Medical students were invited to participate in the course. Before and after VR simulation, participants completed a multiple-choice knowledge assessment, with a maximum score of 10, and a VR experience questionnaire. The primary endpoint compared pre- and post-VR simulation test scores. Secondary endpoints included usability and factors that could affect learning growth and test results. RESULTS: Data from 117 of the 135 participating students was available for analysis. Student test scores improved by an average of 3.4 points (95% CI 3.1-3.7, P < 0.001) after VR course. The secondary endpoints of gender, age, prior knowledge regarding the medical subject, professional training completed in the medical field, video game play, three-dimensional imagination skills, or cyber-sickness had no major impact on test scores or final ranking (top or bottom 25%). The 27 students whose post-VR simulation test scores ranked in the top 25% had no prior experience with VR. The average System Usability Scale score was 86.1, which corresponds to an excellent outcome for user-friendliness. Questionnaire responses post-VR simulation show students (81.2% [95/117]) interest in more VR options in medical school. CONCLUSIONS: We present a freely available software that allows for the development of VR teaching lessons without programming skills.

17.
bioRxiv ; 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38370761

ABSTRACT

Phage therapy is a therapeutic approach to treat multidrug resistant infections that employs lytic bacteriophages (phages) to eliminate bacteria. Despite the abundant evidence for its success as an antimicrobial in Eastern Europe, there is scarce data regarding its effects on the human host. Here, we aimed to understand how lytic phages interact with cells of the airway epithelium, the tissue site that is colonized by bacterial biofilms in numerous chronic respiratory disorders. We determined that interactions between phages and epithelial cells depend on specific phage properties as well as physiochemical features of the microenvironment. Although poor at internalizing phages, the airway epithelium responds to phage exposure by changing its transcriptional profile and secreting antiviral and proinflammatory cytokines that correlate with specific phage families. Overall, our findings indicate that mammalian responses to phages are heterogenous and could potentially alter the way that respiratory local defenses aid in bacterial clearance during phage therapy. Thus, besides phage receptor specificity in a particular bacterial isolate, the criteria to select lytic phages for therapy should be expanded to include mammalian cell responses.

18.
Sci Rep ; 14(1): 2657, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38302552

ABSTRACT

Bacteriophage therapy is one potential strategy to treat antimicrobial resistant or persistent bacterial infections, and the year 2021 marked the centennial of Felix d'Hérelle's first publication on the clinical applications of phages. At the Center for Phage Biology & Therapy at Yale University, a preparatory modular approach has been established to offer safe and potent phages for single-patient investigational new drug applications while recognizing the time constraints imposed by infection(s). This study provides a practical walkthrough of the pipeline with an Autographiviridae phage targeting Pseudomonas aeruginosa (phage vB_PaeA_SB, abbreviated to ΦSB). Notably, a thorough phage characterization and the evolutionary selection pressure exerted on bacteria by phages, analogous to antibiotics, are incorporated into the pipeline.


Subject(s)
Bacteriophages , Phage Therapy , Pseudomonas Infections , Pseudomonas Phages , Humans , Pseudomonas aeruginosa , Universities , Pseudomonas Phages/genetics , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology
19.
J Biol Chem ; 287(50): 41991-2000, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23086930

ABSTRACT

Dysregulated amphiregulin (AR) expression and EGR receptor (EGFR) activation have been described in animal models of pulmonary fibrosis and in patients with idiopathic pulmonary fibrosis. However, the exact role of AR in the pathogenesis of pulmonary fibrosis has not been clearly defined. Here, we show that a potent profibrogenic cytokine TGF-ß1 significantly induced the expression of AR in lung fibroblasts in vitro and in murine lungs in vivo. AR stimulated NIH3T3 fibroblast cell proliferation in a dose-dependent manner. Silencing of AR expression by siRNA or chemical inhibition of EGFR signaling, utilizing AG1478 and gefitinib, significantly reduced the ability of TGF-ß1 to stimulate fibroblast proliferation and expression of α-smooth muscle actin, collagen, and other extracellular matrix-associated genes. TGF-ß1-stimulated activation of Akt, ERK, and Smad signaling was also significantly inhibited by these interventions. Consistent with these in vitro findings, AR expression was impressively increased in the lungs of TGF-ß1 transgenic mice, and either siRNA silencing of AR or chemical inhibition of EGFR signaling significantly reduced TGF-ß1-stimulated collagen accumulation in the lung. These studies showed a novel regulatory role for AR in the pathogenesis of TGF-ß1-induced pulmonary fibrosis. In addition, these studies suggest that AR, or AR-activated EGFR signaling, is a potential therapeutic target for idiopathic pulmonary fibrosis associated with TGF-ß1 activation.


Subject(s)
ErbB Receptors/metabolism , Fibroblasts/metabolism , Glycoproteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Lung/metabolism , MAP Kinase Signaling System , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta1/metabolism , Amphiregulin , Animals , Cell Proliferation , EGF Family of Proteins , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/pathology , Gene Expression Regulation/genetics , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Ligands , Lung/pathology , Mice , Mice, Transgenic , NIH 3T3 Cells , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/genetics
20.
Clin Transplant ; 27(1): E64-71, 2013.
Article in English | MEDLINE | ID: mdl-23278569

ABSTRACT

BACKGROUND: Community acquired respiratory virus (CARV) infections in lung transplant recipients (LTR) have been associated with adverse outcomes, including acute rejection (AR) and decline in allograft function, in some but not in all studies. METHODS: Spirometry and transbronchial biopsy results of LTR diagnosed with CARV infection over a two-yr period were extracted from clinical records. Primary outcomes, studied at 1-2.5 months postinfection, were as follows: (i) incidence of biopsy-proven AR (grade >A0) and (ii) allograft function, defined by forced expiratory volume in one s (FEV(1)). A reference group of biopsies (n = 526) collected during the study period established the baseline incidence of AR. Rhinovirus (RV) and non-rhinovirus (non-RV) infections were analyzed as subgroups. RESULTS: Eighty-seven cases of CARV infection were identified in 59 subjects. Incidences of AR were similar in the post-CARV and reference groups and did not differ significantly after RV vs. non-RV infection. Allograft function declined significantly after non-RV infection, but not after RV infection. CONCLUSIONS: In LTR, CARV infections other than RV are associated with allograft dysfunction at 1-2.5 months after infection. However, CARVs do not appear associated with AR at this time point. The impact of specific CARVs on lung allografts, including the development of chronic allograft rejection, merits further study.


Subject(s)
Graft Rejection/etiology , Lung Transplantation/adverse effects , Picornaviridae Infections/etiology , Respiratory Tract Infections/etiology , DNA, Viral/genetics , Female , Follow-Up Studies , Graft Rejection/epidemiology , Graft Rejection/mortality , Humans , Male , Middle Aged , Picornaviridae Infections/epidemiology , Picornaviridae Infections/mortality , Polymerase Chain Reaction , Prognosis , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/mortality , Rhinovirus/genetics , Rhinovirus/pathogenicity , Spirometry , Survival Rate , Transplantation, Homologous , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL