Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.534
Filter
1.
PLoS Pathog ; 20(7): e1012084, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38976749

ABSTRACT

Little is known about oxygen utilization during infection by bacterial respiratory pathogens. The classical Bordetella species, including B. pertussis, the causal agent of human whooping cough, and B. bronchiseptica, which infects nearly all mammals, are obligate aerobes that use only oxygen as the terminal electron acceptor for electron transport-coupled oxidative phosphorylation. B. bronchiseptica, which occupies many niches, has eight distinct cytochrome oxidase-encoding loci, while B. pertussis, which evolved from a B. bronchiseptica-like ancestor but now survives exclusively in and between human respiratory tracts, has only three functional cytochrome oxidase-encoding loci: cydAB1, ctaCDFGE1, and cyoABCD1. To test the hypothesis that the three cytochrome oxidases encoded within the B. pertussis genome represent the minimum number and class of cytochrome oxidase required for respiratory infection, we compared B. bronchiseptica strains lacking one or more of the eight possible cytochrome oxidases in vitro and in vivo. No individual cytochrome oxidase was required for growth in ambient air, and all three of the cytochrome oxidases conserved in B. pertussis were sufficient for growth in ambient air and low oxygen. Using a high-dose, large-volume persistence model and a low-dose, small-volume establishment of infection model, we found that B. bronchiseptica producing only the three B. pertussis-conserved cytochrome oxidases was indistinguishable from the wild-type strain for infection. We also determined that CyoABCD1 is sufficient to cause the same level of bacterial burden in mice as the wild-type strain and is thus the primary cytochrome oxidase required for murine infection, and that CydAB1 and CtaCDFGE1 fulfill auxiliary roles or are important for aspects of infection we have not assessed, such as transmission. Our results shed light on the environment at the surface of the ciliated epithelium, respiration requirements for bacteria that colonize the respiratory tract, and the evolution of virulence in bacterial pathogens.


Subject(s)
Bordetella Infections , Electron Transport Complex IV , Animals , Mice , Electron Transport Complex IV/metabolism , Electron Transport Complex IV/genetics , Bordetella Infections/microbiology , Respiratory Tract Infections/microbiology , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/metabolism , Bordetella bronchiseptica/enzymology , Humans , Respiratory System/microbiology , Respiratory System/metabolism , Biological Evolution , Bordetella/genetics , Bordetella/enzymology , Bordetella pertussis/genetics , Bordetella pertussis/enzymology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
2.
Int J Pharm ; 661: 124408, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38969264

ABSTRACT

This paper presents a numerical investigation to understand the transport and deposition of sprays emitted by an impinging-jet inhaler in the human respiratory tract under different inhalation flow rates. An injection model is used for the numerical simulations considering the spreading angles of the spray in the two directions, which are measured from experiments. The model parameter is adjusted to match the mean droplet size measured in the previous experiment. A time-varying sinusoidal inhalation flow rate is utilized as airflow conditions, which is closer to the actual situation when using an inhaler. The results demonstrate that the inhalation airflow rate significantly affects the spray's transport behavior and deposition results in the respiratory tract. Both excessively high and low inhalation flow rates lead to an increase in deposition in the mouth-throat. A moderate inhalation flow rate reduces throat deposition while maximizing lung deposition. Higher inhalation flow rates enable faster delivery of the droplets to the lungs, whereas lower inhalation flow rates achieve a more uniform deposition over time in the lungs. The amount of deposition in different parts of the lung lobes follows a fixed order. This study provides valuable insights for optimizing the inhalation flow rate conditions of the impinging-jet inhaler for clinical applications.


Subject(s)
Nebulizers and Vaporizers , Humans , Administration, Inhalation , Respiratory System/metabolism , Aerosols , Lung/metabolism , Particle Size , Equipment Design , Models, Biological , Computer Simulation
3.
Curr Opin Immunol ; 87: 102430, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38824869

ABSTRACT

Lambda interferons (IFNλs), also termed type III interferons (IFNs) or interleukins-28/29, have been in the shadow of type I IFNs for a long time. Their common induction mechanisms and signalling cascades with type I IFNs have made difficult the unwinding of their unique nonredundant functions. However, this is now changing with mounting evidence supporting a major role of IFNλs as a specialized antiviral defense system in the body, mediating protection at mucosal barrier surfaces while limiting immunopathology. Here, we review the latest progress on the complex activities of IFNλs in the respiratory tract, focusing on their multiple effects in IFNλ receptor-expressing cells, the modulation of innate and adaptive immune responses in the context of infections and respiratory diseases, and their similarities and differences with type I IFNs. We also discuss their potential in therapeutic applications and the most recent developments in that direction.


Subject(s)
Adaptive Immunity , Immunity, Innate , Interferon Lambda , Interferons , Respiratory System , Humans , Animals , Interferons/metabolism , Interferons/immunology , Respiratory System/immunology , Respiratory System/metabolism , Signal Transduction/immunology , Interferon Type I/metabolism , Interferon Type I/immunology
4.
Biomed Pharmacother ; 177: 116984, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908203

ABSTRACT

The global public health crisis caused by the COVID-19 pandemic has intensified the global concern regarding viral respiratory tract infections. Despite their considerable impact on health, society and the economy, effective management of these conditions remains a significant challenge. Integrating high-throughput analyses is pivotal for early detection, prognostication of adverse outcomes, elucidating pathogenetic pathways and developing therapeutic approaches. In recent years, microRNAs (miRNAs), a subset of small noncoding RNAs (ncRNAs), have emerged as promising tools for molecular phenotyping. Current evidence suggests that miRNAs could serve as innovative biological markers, aiding in informed medical decision-making. The cost-effective quantification of miRNAs in standardized samples using techniques routinely employed in clinical laboratories has become feasible. In this context, samples obtained from the airways represent a valuable source of information due to their direct exposure to the infectious agent and host response within the respiratory tract. This review explores viral and host miRNA profiling in airway-derived biosamples as a source of molecular information to guide patient management, with a specific emphasis on SARS-CoV-2 infection.


Subject(s)
Biomarkers , COVID-19 , MicroRNAs , Respiratory Tract Infections , SARS-CoV-2 , Humans , MicroRNAs/genetics , COVID-19/genetics , COVID-19/virology , COVID-19/diagnosis , Respiratory Tract Infections/virology , Respiratory Tract Infections/genetics , Respiratory Tract Infections/diagnosis , Biomarkers/metabolism , SARS-CoV-2/genetics , RNA, Viral/genetics , Respiratory System/virology , Respiratory System/metabolism
5.
Exp Physiol ; 109(8): 1370-1384, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38867461

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterised by respiratory muscle injury, inflammation, fibrosis and weakness, ultimately culminating in respiratory failure. The dystrophin-deficient mouse model of DMD (mdx) shows evidence of respiratory muscle remodelling and dysfunction contributing to impaired respiratory system performance. The antioxidant N-acetylcysteine (NAC) has been shown to exert anti-inflammatory and anti-fibrotic effects leading to improved respiratory muscle performance in a range of animal models of muscle dysfunction, including mdx mice, following short-term administration (2 weeks). We sought to build on previous work by exploring the effects of chronic NAC administration (3 months) on respiratory system performance in mdx mice. One-month-old male mdx mice were randomised to receive normal drinking water (n = 30) or 1% NAC in the drinking water (n = 30) for 3 months. At 4 months of age, we assessed breathing in conscious mice by plethysmography followed by ex vivo assessment of diaphragm force-generating capacity. Additionally, diaphragm histology was performed. In separate studies, in anaesthetised mice, respiratory electromyogram (EMG) activity and inspiratory pressure across a range of behaviours were determined, including assessment of peak inspiratory pressure-generating capacity. NAC treatment did not affect force-generating capacity of the mdx diaphragm. Collagen content and immune cell infiltration were unchanged in mdx + NAC compared with mdx diaphragms. Additionally, there was no significant effect of NAC on breathing, ventilatory responsiveness, inspiratory EMG activity or inspiratory pressure across the range of behaviours from basal conditions to peak system performance. We conclude that chronic NAC treatment has no apparent beneficial effects on respiratory system performance in the mdx mouse model of DMD suggesting limited potential of NAC treatment alone for human DMD.


Subject(s)
Acetylcysteine , Diaphragm , Disease Models, Animal , Mice, Inbred mdx , Muscular Dystrophy, Duchenne , Animals , Acetylcysteine/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/physiopathology , Male , Mice , Diaphragm/drug effects , Diaphragm/physiopathology , Mice, Inbred C57BL , Respiratory Muscles/drug effects , Respiratory Muscles/physiopathology , Respiration/drug effects , Antioxidants/pharmacology , Respiratory System/drug effects , Respiratory System/physiopathology , Respiratory System/metabolism
6.
Methods Mol Biol ; 2820: 165-185, 2024.
Article in English | MEDLINE | ID: mdl-38941023

ABSTRACT

The upper respiratory tract (URT) is home to a diverse range of microbial species. Respiratory infections disturb the microbial flora in the URT, putting people at risk of secondary infections. The potential dangers and clinical effects of bacterial and fungal coinfections with SARS-CoV-2 support the need to investigate the microbiome of the URT using clinical samples. Mass spectrometry (MS)-based metaproteomics analysis of microbial proteins is a novel approach to comprehensively assess the clinical specimens with complex microbial makeup. The coronavirus that causes severe acute respiratory syndrome (SARS-CoV-2) is responsible for the COVID-19 pandemic resulting in a plethora of microbial coinfections impeding therapy, prognosis, and overall disease management. In this chapter, the corresponding workflows for MS-based shotgun proteomics and metaproteomic analysis are illustrated.


Subject(s)
COVID-19 , Coinfection , Proteomics , SARS-CoV-2 , Humans , COVID-19/virology , COVID-19/complications , Proteomics/methods , Coinfection/microbiology , Coinfection/virology , SARS-CoV-2/isolation & purification , Microbiota , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Respiratory Tract Infections/diagnosis , Mass Spectrometry/methods , Proteome/analysis , Respiratory System/microbiology , Respiratory System/metabolism , Respiratory System/virology
7.
Respir Physiol Neurobiol ; 326: 104271, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38703974

ABSTRACT

The objective of this study is to explore the transport, size growth, and deposition of Salbutamol Sulphate (SS) using Computational Fluid Dynamics (CFD). A CT-based realistic model of human airways from the oral cavity to the 5th generation of the lung was utilized as the computational domain. Four Test Cases (TC) with varying temperature and relative humidity (RH) under two inspiratory waveforms were considered to completely evaluate the impact of inhalation conditions on particle growth. Salbutamol Sulphate (SS) is a ß2-adrenergic agonist and has been extensively used for asthma treatment. A monodispersed distribution of SS particles with an initial diameter of 167 nm was considered at the mouth inlet based on pharmaceutical data. Results indicated that inhalation of saturated/supersaturated air (RH>100%) leads to significant hygroscopic growth of SS particles with a factor of 10. In addition, the deposition efficiency of SS particles under the Quick and Deep (QD) inhalation profile was enhanced as the flow temperature and humidity increased. However, the implementation of Slow and Deep (SD) inspiratory waveform revealed that the same particle size growth is achieved in the respiratory system with lower deposition efficiency in the mouth-throat (less than 3%) and tracheobronchial airway (less than 2.18%). For the escaped particles form the right lung, in the SD waveform under TC 3, the maximum particle size distribution was for 600 nm particles with 25% probability. In the left lung, 30% of the particles were increased up to 950 nm in size. For the QD waveform in TC 3 and TC4, the most frequent particles were 800 nm with 36% probability. This holds practical significance in the context of deep lung delivery for asthmatic patients with enhanced deposition efficiency and large particle size. The findings of the present study can contribute to the development of targeted drug delivery strategies for the treatment of pulmonary diseases using hygroscopic dry powder formulations.


Subject(s)
Albuterol , Computer Simulation , Humans , Albuterol/administration & dosage , Albuterol/pharmacology , Administration, Inhalation , Bronchodilator Agents/administration & dosage , Bronchodilator Agents/pharmacology , Hydrodynamics , Models, Biological , Particle Size , Humidity , Wettability , Respiratory System/drug effects , Respiratory System/metabolism , Lung/drug effects , Lung/metabolism
8.
J Vis Exp ; (205)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38587373

ABSTRACT

cAMP Difference Detector In Situ (cADDis) is a novel biosensor that allows for the continuous measurement of cAMP levels in living cells. The biosensor is created from a circularly permuted fluorescent protein linked to the hinge region of Epac2. This creates a single fluorophore biosensor that displays either increased or decreased fluorescence upon binding of cAMP. The biosensor exists in red and green upward versions, as well as green downward versions, and several red and green versions targeted to subcellular locations. To illustrate the effectiveness of the biosensor, the green downward version, which decreases in fluorescence upon cAMP binding, was used. Two protocols using this sensor are demonstrated: one utilizing a 96-well plate reading spectrophotometer compatible with high-throughput screening and another utilizing single-cell imaging on a fluorescent microscope. On the plate reader, HEK-293 cells cultured in 96-well plates were stimulated with 10 µM forskolin or 10 nM isoproterenol, which induced rapid and large decreases in fluorescence in the green downward version. The biosensor was used to measure cAMP levels in individual human airway smooth muscle (HASM) cells monitored under a fluorescent microscope. The green downward biosensor displayed similar responses to populations of cells when stimulated with forskolin or isoproterenol. This single-cell assay allows visualization of the biosensor location at 20x and 40x magnification. Thus, this cAMP biosensor is sensitive and flexible, allowing real-time measurement of cAMP in both immortalized and primary cells, and with single cells or populations of cells. These attributes make cADDis a valuable tool for studying cAMP signaling dynamics in living cells.


Subject(s)
Cyclic AMP , Respiratory System , Humans , Cyclic AMP/metabolism , Isoproterenol/pharmacology , Colforsin/pharmacology , HEK293 Cells , Respiratory System/metabolism
9.
Pharm Dev Technol ; 29(5): 477-481, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38656248

ABSTRACT

This study was the first attempt to visualize pulmonary retention of nanocarriers (NCs) with the use of the P2 probe, a new water-initiated aggregation-caused fluorescent-quenching (ACQ) dye, for the development of NCs with long-lasting retention in the respiratory system (RS). Flash nanoprecipitation was used to fabricate mucopenetrating NCs (MP/NCs) and mucoadhesive NCs (MA/NCs). Both NCs were labeled with the P2 probe, and their distribution and retention in RS were visualized after intratracheal administration to rats. MP/NCs and MA/NCs had a mean diameter below 200 nm and ζ-potential of 0 and 48 mV, respectively. MA/NCs showed three times stronger interactions with mucin than MP/NCs, resulting in significantly lower diffusiveness in mucus. The P2 probe exhibited an ACQ effect with negligible rekindling in simulated lung fluid, and the spectroscopic data suggested applicability to reliable imaging of insufflated NCs. In confocal laser scanning microscopic and in vivo imaging system images of the rat RS, MA/NCs were locally deposited in the respiratory tract and transported toward the pharynx by mucocilliary clearance (MCC). In contrast, MP/NCs diffused in the respiratory mucus were less subject to the influence of MCC. Based on the results from the bioimaging study using the P2 probe, MP/NCs could offer enhanced pulmonary retention of drugs compared with MA/NCs.


Subject(s)
Drug Carriers , Nanoparticles , Animals , Rats , Nanoparticles/chemistry , Drug Carriers/chemistry , Male , Rats, Sprague-Dawley , Respiratory System/metabolism , Respiratory System/drug effects , Fluorescent Dyes/chemistry , Fluorescent Dyes/administration & dosage
10.
Toxicology ; 504: 153781, 2024 May.
Article in English | MEDLINE | ID: mdl-38493948

ABSTRACT

This comprehensive review focuses on various dimensions of nanoparticle toxicity, emphasizing toxicological characteristics, assessment techniques, and examinations of relevant studies on the effects on biological systems. The primary objective is to comprehend the potential risks associated with nanoparticles and to provide efficient strategies for mitigating them by consolidating current research discoveries. For in-depth insights, the discussions extend to crucial aspects such as toxicity associated with different nanoparticles, human exposure, and nanoparticle deposition in the human respiratory tract. The analysis utilizes the multiple-path particle dosimetry (MPPD) modeling for computational simulation. The SiO2 nanoparticles with a volume concentration of 1% and a particle size of 50 nm are used to depict the MPPD modeling of the Left upper (LU), left lower (LL), right upper (RU), right middle (RM), and right lower (RL) lobes in the respiratory tract. The analysis revealed a substantial 67.5% decrease in the deposition fraction as the particle size increased from 10 nm to 100 nm. Graphical representation emphasizes the significant impact of exposure path selection on nanoparticle deposition, with distinct deposition values observed for nasal, oral, oronasal-mouth breather, oronasal - normal augmenter, and endotracheal paths (0.00291 µg, 0.00332 µg, 0.00297 µg, 0.00291 µg, and 0.00383 µg, respectively). Consistent with the focus of the review, the article also addresses crucial mitigation strategies for managing nanoparticle toxicity.


Subject(s)
Nanoparticles , Respiratory System , Humans , Nanoparticles/toxicity , Respiratory System/drug effects , Respiratory System/metabolism , Animals , Risk Factors , Inhalation Exposure/adverse effects , Particle Size , Risk Assessment
11.
Cells ; 13(6)2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38534319

ABSTRACT

Chronic Obstructive Pulmonary Disease (COPD) is a pathological condition of the respiratory system characterized by chronic airflow obstruction, associated with changes in the lung parenchyma (pulmonary emphysema), bronchi (chronic bronchitis) and bronchioles (small airways disease). In the last years, the importance of phenotyping and endotyping COPD patients has strongly emerged. Metabolomics refers to the study of metabolites (both intermediate or final products) and their biological processes in biomatrices. The application of metabolomics to respiratory diseases and, particularly, to COPD started more than one decade ago and since then the number of scientific publications on the topic has constantly grown. In respiratory diseases, metabolomic studies have focused on the detection of metabolites derived from biomatrices such as exhaled breath condensate, bronchoalveolar lavage, and also plasma, serum and urine. Mass Spectrometry and Nuclear Magnetic Resonance Spectroscopy are powerful tools in the precise identification of potentially prognostic and treatment response biomarkers. The aim of this article was to comprehensively review the relevant literature regarding the applications of metabolomics in COPD, clarifying the potential clinical utility of the metabolomic profile from several biologic matrices in detecting biomarkers of disease and prognosis for COPD. Meanwhile, a complete description of the technological instruments and techniques currently adopted in the metabolomics research will be described.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Humans , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory System/metabolism , Metabolomics/methods , Biomarkers/metabolism , Mass Spectrometry/methods
12.
Int J Mol Sci ; 25(3)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38339210

ABSTRACT

The respiratory mucus, a viscoelastic gel, effectuates a primary line of the airway defense when operated by the mucociliary clearance. In chronic respiratory diseases (CRDs), such as asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF), the mucus is overproduced and its solid content augments, changing its structure and viscoelastic properties and determining a derangement of essential defense mechanisms against opportunistic microbial (virus and bacteria) pathogens. This ensues in damaging of the airways, leading to a vicious cycle of obstruction and infection responsible for the harsh clinical evolution of these CRDs. Here, we review the essential features of normal and pathological mucus (i.e., sputum in CF, COPD, and asthma), i.e., mucin content, structure (mesh size), micro/macro-rheology, pH, and osmotic pressure, ending with the awareness that sputum biomarkers (mucins, inflammatory proteins and peptides, and metabolites) might serve to indicate acute exacerbation and response to therapies. There are some indications that old and novel treatments may change the structure, viscoelastic properties, and biomarker content of sputum; however, a wealth of work is still needed to embrace these measures as correlates of disease severity in association with (or even as substitutes of) pulmonary functional tests.


Subject(s)
Asthma , Cystic Fibrosis , Pulmonary Disease, Chronic Obstructive , Respiration Disorders , Humans , Mucus/metabolism , Respiration Disorders/metabolism , Respiratory System/metabolism , Cystic Fibrosis/metabolism , Asthma/metabolism , Sputum/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Mucins/metabolism
13.
Life Sci Alliance ; 7(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38176727

ABSTRACT

Activating transcription factor 4 (Atf4), which is modulated by the protein kinase RNA-like ER kinase (PERK), is a stress-induced transcription factor responsible for controlling the expression of a wide range of adaptive genes, enabling cells to withstand stressful conditions. However, the impact of the Atf4 signaling pathway on airway regeneration remains poorly understood. In this study, we used mouse airway epithelial cell culture models to investigate the role of PERK/Atf4 in respiratory tract differentiation. Through pharmacological inhibition and silencing of ATF4, we uncovered the crucial involvement of PERK/Atf4 in the differentiation of basal stem cells, leading to a reduction in the number of secretory cells. ChIP-seq analysis revealed direct binding of ATF4 to regulatory elements of genes associated with osteoblast differentiation and secretory cell function. Our findings provide valuable insights into the role of ATF4 in airway epithelial differentiation and its potential involvement in innate immune responses and cellular adaptation to stress.


Subject(s)
Endoplasmic Reticulum Stress , eIF-2 Kinase , Animals , Mice , eIF-2 Kinase/genetics , Endoplasmic Reticulum Stress/genetics , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Cell Differentiation/genetics , Respiratory System/metabolism
14.
J Virol ; 97(10): e0127123, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37819131

ABSTRACT

IMPORTANCE: The respiratory tract of humans is constantly exposed to potentially harmful agents, such as small particles or pathogens, and thus requires protective measures. Respiratory mucus that lines the airway epithelia plays a major role in the prevention of viral infections by limiting the mobility of viruses, allowing subsequent mucociliary clearance. Understanding the interplay between respiratory mucus and viruses can help elucidate host and virus characteristics that enable the initiation of infection. Here, we tested a panel of primary influenza A viruses of avian or human origin for their sensitivity to mucus derived from primary human airway cultures and found that differences between virus strains can be mapped to viral neuraminidase activity. We also show that binding of influenza A viruses to decoy receptors on highly glycosylated mucus components constitutes the major inhibitory function of mucus against influenza A viruses.


Subject(s)
Influenza A virus , Influenza, Human , Mucus , Neuraminidase , Animals , Humans , Birds , Influenza A virus/metabolism , Mucus/metabolism , Neuraminidase/metabolism , Respiratory System/metabolism
15.
Radiat Prot Dosimetry ; 199(15-16): 1838-1843, 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37819295

ABSTRACT

The Human Respiratory Tract Model described in Publication 130 of the International Commission on Radiological Protection provides some mechanisms to account for retention of material that can be subject to little to no mechanical transport or absorption into the blood. One of these mechanisms is 'binding', which refers to a process by which a fraction ('bound fraction') of the dissolved material chemically binds to the tissue of the airway wall. The value of the bound fraction can have a significant impact on the radiation doses imparted to different parts of the respiratory tract. To properly evaluate-and quantify-bound fraction for an element, one would need information on long-term retention of the element in individual compartments of the respiratory tract. Such data on regional retention of plutonium in the respiratory tract of four workers-who had inhaled materials with solubility ranging from soluble nitrate to very insoluble high-fired oxides-were obtained at the United States Transuranium and Uranium Registries. An assumption of bound fraction alone was found to be inconsistent with this dataset and also with a review of the literature. Several studies show evidence of retention of a large amount of Pu activity in the scar tissues of humans and experimental animals, and accordingly, a model structure with scar-tissue compartments was proposed. The transfer rates to these compartments were determined using Markov Chain Monte Carlo analysis of the bioassay and post-mortem data, considering the uncertainties associated with deposition, dissolution and particle clearance parameters. The models predicted that a significant amount-between 20 and 100% for the cases analyzed-of plutonium retained in the respiratory tract was sequestered in the scar tissues. Unlike chemically-bound Pu that irradiates sensitive epithelial cells, Pu in scar tissues may not be dosimetrically significant because the scar tissues absorb most, if not all, of the energy from alpha emissions.


Subject(s)
Plutonium , Animals , Humans , United States , Plutonium/analysis , Radiation Dosage , Cicatrix/metabolism , Models, Biological , Respiratory System/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 325(6): L765-L775, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37847709

ABSTRACT

Airway mucociliary clearance (MCC) is required for host defense and is often diminished in chronic lung diseases. Effective clearance depends upon coordinated actions of the airway epithelium and a mobile mucus layer. Dysregulation of the primary secreted airway mucin proteins, MUC5B and MUC5AC, is associated with a reduction in the rate of MCC; however, how other secreted proteins impact the integrity of the mucus layer and MCC remains unclear. We previously identified the gene Bpifb1/Lplunc1 as a regulator of airway MUC5B protein levels using genetic approaches. Here, we show that BPIFB1 is required for effective MCC in vivo using Bpifb1 knockout (KO) mice. Reduced MCC in Bpifb1 KO mice occurred in the absence of defects in epithelial ion transport or reduced ciliary beat frequency. Loss of BPIFB1 in vivo and in vitro altered biophysical and biochemical properties of mucus that have been previously linked to impaired MCC. Finally, we detected colocalization of BPIFB1 and MUC5B in secretory granules in mice and the protein mesh of secreted mucus in human airway epithelia cultures. Collectively, our findings demonstrate that BPIFB1 is an important component of the mucociliary apparatus in mice and a key component of the mucus protein network.NEW & NOTEWORTHY BPIFB1, also known as LPLUNC1, was found to regulate mucociliary clearance (MCC), a key aspect of host defense in the airway. Loss of this protein was also associated with altered biophysical and biochemical properties of mucus that have been previously linked to impaired MCC.


Subject(s)
Lung Diseases , Mucociliary Clearance , Mice , Humans , Animals , Mucociliary Clearance/physiology , Respiratory System/metabolism , Mucus/metabolism , Lung Diseases/metabolism , Mice, Knockout
17.
Expert Rev Respir Med ; 17(10): 903-917, 2023.
Article in English | MEDLINE | ID: mdl-37905552

ABSTRACT

INTRODUCTION: Asthma is a chronic lung disease influenced by environmental and inflammatory triggers and involving complex signaling pathways across resident airway cells such as epithelium, airway smooth muscle, fibroblasts, and immune cells. While our understanding of asthma pathophysiology is continually progressing, there is a growing realization that cellular microdomains play critical roles in mediating signaling relevant to asthma in the context of contractility and remodeling. Mechanosensitive pathways are increasingly recognized as important to microdomain signaling, with Piezo and transient receptor protein (TRP) channels at the plasma membrane considered important for converting mechanical stimuli into cellular behavior. Given their ion channel properties, particularly Ca2+ conduction, a question becomes whether and how mechanosensitive channels contribute to Ca2+ microdomains in airway cells relevant to asthma. AREAS COVERED: Mechanosensitive TRP and Piezo channels regulate key Ca2+ regulatory proteins such as store operated calcium entry (SOCE) involving STIM and Orai channels, and sarcoendoplasmic (SR) mechanisms such as IP3 receptor channels (IP3Rs), and SR Ca2+ ATPase (SERCA) that are important in asthma pathophysiology including airway hyperreactivity and remodeling. EXPERT OPINION: Physical and/or functional interactions between Ca2+ regulatory proteins and mechanosensitive channels such as TRP and Piezo can toward understanding asthma pathophysiology and identifying novel therapeutic approaches.


Subject(s)
Asthma , Calcium , Humans , Calcium/metabolism , Calcium Signaling/physiology , Respiratory System/metabolism , Cell Membrane/metabolism , Asthma/metabolism
18.
Nature ; 619(7969): 338-347, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380775

ABSTRACT

Spillover events of avian influenza A viruses (IAVs) to humans could represent the first step in a future pandemic1. Several factors that limit the transmission and replication of avian IAVs in mammals have been identified. There are several gaps in our understanding to predict which virus lineages are more likely to cross the species barrier and cause disease in humans1. Here, we identified human BTN3A3 (butyrophilin subfamily 3 member A3)2 as a potent inhibitor of avian IAVs but not human IAVs. We determined that BTN3A3 is expressed in human airways and its antiviral activity evolved in primates. We show that BTN3A3 restriction acts primarily at the early stages of the virus life cycle by inhibiting avian IAV RNA replication. We identified residue 313 in the viral nucleoprotein (NP) as the genetic determinant of BTN3A3 sensitivity (313F or, rarely, 313L in avian viruses) or evasion (313Y or 313V in human viruses). However, avian IAV serotypes, such as H7 and H9, that spilled over into humans also evade BTN3A3 restriction. In these cases, BTN3A3 evasion is due to substitutions (N, H or Q) in NP residue 52 that is adjacent to residue 313 in the NP structure3. Thus, sensitivity or resistance to BTN3A3 is another factor to consider in the risk assessment of the zoonotic potential of avian influenza viruses.


Subject(s)
Birds , Host Microbial Interactions , Influenza A virus , Influenza in Birds , Influenza, Human , Viral Zoonoses , Animals , Humans , Birds/virology , Influenza A virus/classification , Influenza A virus/genetics , Influenza A virus/growth & development , Influenza A virus/isolation & purification , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza, Human/prevention & control , Influenza, Human/transmission , Influenza, Human/virology , Primates , Respiratory System/metabolism , Respiratory System/virology , Risk Assessment , Viral Zoonoses/prevention & control , Viral Zoonoses/transmission , Viral Zoonoses/virology , Virus Replication
19.
Math Med Biol ; 40(3): 238-265, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37285178

ABSTRACT

Excessive activation of the regulatory cytokine transforming growth factor $\beta $ (TGF-$\beta $) via contraction of airway smooth muscle (ASM) is associated with the development of asthma. In this study, we develop an ordinary differential equation model that describes the change in density of the key airway wall constituents, ASM and extracellular matrix (ECM), and their interplay with subcellular signalling pathways leading to the activation of TGF-$\beta $. We identify bistable parameter regimes where there are two positive steady states, corresponding to either reduced or elevated TGF-$\beta $ concentration, with the latter leading additionally to increased ASM and ECM density. We associate the former with a healthy homeostatic state and the latter with a diseased (asthmatic) state. We demonstrate that external stimuli, inducing TGF-$\beta $ activation via ASM contraction (mimicking an asthmatic exacerbation), can perturb the system irreversibly from the healthy state to the diseased one. We show that the properties of the stimuli, such as their frequency or strength, and the clearance of surplus active TGF-$\beta $, are important in determining the long-term dynamics and the development of disease. Finally, we demonstrate the utility of this model in investigating temporal responses to bronchial thermoplasty, a therapeutic intervention in which ASM is ablated by applying thermal energy to the airway wall. The model predicts the parameter-dependent threshold damage required to obtain irreversible reduction in ASM content, suggesting that certain asthma phenotypes are more likely to benefit from this intervention.


Subject(s)
Asthma , Respiratory System , Humans , Respiratory System/metabolism , Asthma/genetics , Asthma/metabolism , Muscle, Smooth/metabolism , Extracellular Matrix/metabolism , Transforming Growth Factor beta/metabolism
20.
Glycobiology ; 33(6): 476-489, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37115803

ABSTRACT

The COVID-19 global pandemic has underscored the need to understand how viruses and other pathogens are able to infect and replicate within the respiratory system. Recent studies have highlighted the role of highly O-glycosylated mucins in the protection of the respiratory system as well as how mucin-type O-glycosylation may be able to modify viral infectivity. Therefore, we set out to identify the specific genes controlling mucin-type O-glycosylation throughout the mouse respiratory system as well as determine how their expression and the expression of respiratory mucins is influenced by infection or injury. Here, we show that certain mucins and members of the Galnt family are abundantly expressed in specific respiratory tissues/cells and demonstrate unique patterns of O-glycosylation across diverse respiratory tissues. Moreover, we find that the expression of certain Galnts and mucins is altered during lung infection and injury in experimental mice challenged with infectious agents, toxins, and allergens. Finally, we examine gene expression changes of Galnts and mucins in a mouse model of SARS-CoV-2 infection. Our work provides foundational knowledge regarding the specific expression of Galnt enzyme family members and mucins throughout the respiratory system, and how their expression is altered upon lung infection and injury.


Subject(s)
COVID-19 , Mucins , Animals , Mice , Mucins/genetics , Mucins/metabolism , Glycosylation , COVID-19/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Respiratory System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL