Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Publication year range
1.
Physiol Rev ; 100(3): 1065-1075, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32216698

ABSTRACT

Patients with hypertension, diabetes, coronary heart disease, cerebrovascular illness, chronic obstructive pulmonary disease, and kidney dysfunction have worse clinical outcomes when infected with SARS-CoV-2, for unknown reasons. The purpose of this review is to summarize the evidence for the existence of elevated plasmin(ogen) in COVID-19 patients with these comorbid conditions. Plasmin, and other proteases, may cleave a newly inserted furin site in the S protein of SARS-CoV-2, extracellularly, which increases its infectivity and virulence. Hyperfibrinolysis associated with plasmin leads to elevated D-dimer in severe patients. The plasmin(ogen) system may prove a promising therapeutic target for combating COVID-19.


Subject(s)
Coronavirus Infections/blood , Disease Susceptibility/blood , Fibrinolysin/metabolism , Host-Pathogen Interactions/physiology , Plasminogen/metabolism , Pneumonia, Viral/blood , Betacoronavirus/metabolism , Betacoronavirus/pathogenicity , COVID-19 , Comorbidity , Coronavirus Infections/epidemiology , Coronavirus Infections/mortality , Coronavirus Infections/physiopathology , Humans , Pandemics , Pneumonia, Viral/epidemiology , Pneumonia, Viral/mortality , Pneumonia, Viral/physiopathology , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/physiopathology , Respiratory Distress Syndrome/virology , Risk Factors , SARS-CoV-2
2.
Am J Physiol Lung Cell Mol Physiol ; 326(5): L596-L603, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38469648

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a fatal pulmonary disorder characterized by severe hypoxia and inflammation. ARDS is commonly triggered by systemic and pulmonary infections, with bacteria and viruses. Notable pathogens include Pseudomonas aeruginosa, Streptococcus aureus, Enterobacter species, coronaviruses, influenza viruses, and herpesviruses. COVID-19 ARDS represents the latest etiological phenotype of the disease. The pathogenesis of ARDS caused by bacteria and viruses exhibits variations in host immune responses and lung mesenchymal injury. We postulate that the systemic and pulmonary metabolomics profiles of ARDS induced by COVID-19 pathogens may exhibit distinctions compared with those induced by other infectious agents. This review aims to compare metabolic signatures in blood and lung specimens specifically within the context of ARDS. Both prevalent and phenotype-specific metabolomic signatures, including but not limited to glycolysis, ketone body production, lipid oxidation, and dysregulation of the kynurenine pathways, were thoroughly examined in this review. The distinctions in metabolic signatures between COVID-19 and non-COVID ARDS have the potential to reveal new biomarkers, elucidate pathogenic mechanisms, identify druggable targets, and facilitate differential diagnosis in the future.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , SARS-CoV-2 , Humans , COVID-19/metabolism , COVID-19/complications , COVID-19/virology , COVID-19/pathology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/virology , SARS-CoV-2/metabolism , Lung/metabolism , Lung/virology , Lung/pathology , Metabolome , Biomarkers/metabolism , Biomarkers/blood , Metabolomics/methods
3.
PLoS Comput Biol ; 18(10): e1010603, 2022 10.
Article in English | MEDLINE | ID: mdl-36269761

ABSTRACT

Metaproteomics based on high-throughput tandem mass spectrometry (MS/MS) plays a crucial role in characterizing microbiome functions. The acquired MS/MS data is searched against a protein sequence database to identify peptides, which are then used to infer a list of proteins present in a metaproteome sample. While the problem of protein inference has been well-studied for proteomics of single organisms, it remains a major challenge for metaproteomics of complex microbial communities because of the large number of degenerate peptides shared among homologous proteins in different organisms. This challenge calls for improved discrimination of true protein identifications from false protein identifications given a set of unique and degenerate peptides identified in metaproteomics. MetaLP was developed here for protein inference in metaproteomics using an integrative linear programming method. Taxonomic abundance information extracted from metagenomics shotgun sequencing or 16s rRNA gene amplicon sequencing, was incorporated as prior information in MetaLP. Benchmarking with mock, human gut, soil, and marine microbial communities demonstrated significantly higher numbers of protein identifications by MetaLP than ProteinLP, PeptideProphet, DeepPep, PIPQ, and Sipros Ensemble. In conclusion, MetaLP could substantially improve protein inference for complex metaproteomes by incorporating taxonomic abundance information in a linear programming model.


Subject(s)
Programming, Linear , Tandem Mass Spectrometry , Humans , RNA, Ribosomal, 16S/genetics , Proteins/chemistry , Peptides/chemistry
4.
Am J Physiol Lung Cell Mol Physiol ; 323(5): L515-L524, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36098461

ABSTRACT

Failure to regenerate injured alveoli functionally and promptly causes a high incidence of fatality in coronavirus disease 2019 (COVID-19). How elevated plasminogen activator inhibitor-1 (PAI-1) regulates the lineage of alveolar type 2 (AT2) cells for re-alveolarization has not been studied. This study aimed to examine the role of PAI-1-Wnt5a-ß catenin cascades in AT2 fate. Dramatic reduction in AT2 yield was observed in Serpine1Tg mice. Elevated PAI-1 level suppressed organoid number, development efficiency, and total surface area in vitro. Anti-PAI-1 neutralizing antibody restored organoid number, proliferation and differentiation of AT2 cells, and ß-catenin level in organoids. Both Wnt family member 5A (Wnt5a) and Wnt5a-derived N-butyloxycarbonyl hexapeptide (Box5) altered the lineage of AT2 cells. This study demonstrates that elevated PAI-1 regulates AT2 proliferation and differentiation via the Wnt5a/ß catenin cascades. PAI-1 could serve as autocrine signaling for lung injury repair.


Subject(s)
COVID-19 , Plasminogen Activator Inhibitor 1 , Wnt-5a Protein , beta Catenin , Animals , Mice , Antibodies, Neutralizing , beta Catenin/metabolism , Down-Regulation , Wnt Signaling Pathway/physiology , Wnt-5a Protein/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Pulmonary Alveoli/cytology , Cell Proliferation
5.
Respir Res ; 21(1): 132, 2020 May 29.
Article in English | MEDLINE | ID: mdl-32471489

ABSTRACT

BACKGROUND: Chronic tissue injury was shown to induce progressive scarring in fibrotic diseases such as idiopathic pulmonary fibrosis (IPF), while an array of repair/regeneration and stress responses come to equilibrium to determine the outcome of injury at the organ level. In the lung, type I alveolar epithelial (ATI) cells constitute the epithelial barrier, while type II alveolar epithelial (ATII) cells play a pivotal role in regenerating the injured distal lungs. It had been demonstrated that eukaryotic cells possess repair machinery that can quickly patch the damaged plasma membrane after injury, and our previous studies discovered the membrane-mending role of Tripartite motif containing 72 (TRIM72) that expresses in a limited number of tissues including the lung. Nevertheless, the role of alveolar epithelial cell (AEC) repair in the pathogenesis of IPF has not been examined yet. METHOD: In this study, we tested the specific roles of TRIM72 in the repair of ATII cells and the development of lung fibrosis. The role of membrane repair was accessed by saponin assay on isolated primary ATII cells and rat ATII cell line. The anti-fibrotic potential of TRIM72 was tested with bleomycin-treated transgenic mice. RESULTS: We showed that TRIM72 was upregulated following various injuries and in human IPF lungs. However, TRIM72 expression in ATII cells of the IPF lungs had aberrant subcellular localization. In vitro studies showed that TRIM72 repairs membrane injury of immortalized and primary ATIIs, leading to inhibition of stress-induced p53 activation and reduction in cell apoptosis. In vivo studies demonstrated that TRIM72 protects the integrity of the alveolar epithelial layer and reduces lung fibrosis. CONCLUSION: Our results suggest that TRIM72 protects injured lungs and ameliorates fibrosis through promoting post-injury repair of AECs.


Subject(s)
Alveolar Epithelial Cells/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/prevention & control , Lung/metabolism , Tripartite Motif Proteins/biosynthesis , Alveolar Epithelial Cells/drug effects , Animals , Bleomycin/toxicity , Female , HEK293 Cells , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Lung/drug effects , Male , Mice , Mice, 129 Strain , Mice, Knockout , Recombinant Proteins/biosynthesis
6.
Cell Biol Toxicol ; 36(6): 571-589, 2020 12.
Article in English | MEDLINE | ID: mdl-32588239

ABSTRACT

Smoke inhalation injury is the leading cause of death in firefighters and victims. Inhaled hot air and toxic smoke are the predominant hazards to the respiratory epithelium. We aimed to analyze the effects of thermal stress and smoke aldehyde on the permeability of the airway epithelial barrier. Transepithelial resistance (RTE) and short-circuit current (ISC) of mouse tracheal epithelial monolayers were digitized by an Ussing chamber setup. Zonula occludens-1 tight junctions were visualized under confocal microscopy. A cell viability test and fluorescein isothiocyanate-dextran assay were performed. Thermal stress (40 °C) decreased RTE in a two-phase manner. Meanwhile, thermal stress increased ISC followed by its decline. Na+ depletion, amiloride (an inhibitor for epithelial Na+ channels [ENaCs]), ouabain (a blocker for Na+/K+-ATPase), and CFTRinh-172 (a blocker of cystic fibrosis transmembrane regulator [CFTR]) altered the responses of RTE and ISC to thermal stress. Steady-state 40 °C increased activity of ENaCs, Na+/K+-ATPase, and CFTR. Acrolein, one of the main oxidative unsaturated aldehydes in fire smoke, eliminated RTE and ISC. Na+ depletion, amiloride, ouabain, and CFTRinh-172 suppressed acrolein-sensitive ISC, but showed activating effects on acrolein-sensitive RTE. Thermal stress or acrolein disrupted zonula occludens-1 tight junctions, increased fluorescein isothiocyanate-dextran permeability but did not cause cell death or detachment. The synergistic effects of thermal stress and acrolein exacerbated the damage to monolayers. In conclusion, the paracellular pathway mediated by the tight junctions and the transcellular pathway mediated by active and passive ion transport pathways contribute to impairment of the airway epithelial barrier caused by thermal stress and acrolein. Graphical abstract Thermal stress and acrolein are two essential determinants for smoke inhalation injury, impairing airway epithelial barrier. Transcellular ion transport pathways via the ENaC, CFTR, and Na/K-ATPase are interrupted by both thermal stress and acrolein, one of the most potent smoke toxins. Heat and acrolein damage the integrity of the airway epithelium through suppressing and relocating the tight junctions.


Subject(s)
Acrolein/toxicity , Bronchi/drug effects , Epithelial Cells/drug effects , Hot Temperature/adverse effects , Membrane Transport Proteins/metabolism , Smoke Inhalation Injury/etiology , Smoke/adverse effects , Trachea/drug effects , Animals , Bronchi/metabolism , Bronchi/pathology , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Electric Impedance , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Sodium Channels/metabolism , Female , Humans , Inhalation Exposure/adverse effects , Ion Transport , Male , Mice, Inbred C57BL , Permeability , Smoke Inhalation Injury/metabolism , Smoke Inhalation Injury/pathology , Sodium-Potassium-Exchanging ATPase/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/pathology , Trachea/metabolism , Trachea/pathology , Zonula Occludens-1 Protein/metabolism
7.
J Med Genet ; 55(3): 143-149, 2018 03.
Article in English | MEDLINE | ID: mdl-29301855

ABSTRACT

Respiratory diseases, which are leading causes of mortality and morbidity in the world, are dysfunctions of the nasopharynx, the trachea, the bronchus, the lung and the pleural cavity. Symptoms of chronic respiratory diseases, such as cough, sneezing and difficulty breathing, may seriously affect the productivity, sleep quality and physical and mental well-being of patients, and patients with acute respiratory diseases may have difficulty breathing, anoxia and even life-threatening respiratory failure. Respiratory diseases are generally heterogeneous, with multifaceted causes including smoking, ageing, air pollution, infection and gene mutations. Clinically, a single pulmonary disease can exhibit more than one phenotype or coexist with multiple organ disorders. To correct abnormal function or repair injured respiratory tissues, one of the most promising techniques is to correct mutated genes by gene editing, as some gene mutations have been clearly demonstrated to be associated with genetic or heterogeneous respiratory diseases. Zinc finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN) and clustered regulatory interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) systems are three innovative gene editing technologies developed recently. In this short review, we have summarised the structure and operating principles of the ZFNs, TALENs and CRISPR/Cas9 systems and their preclinical and clinical applications in respiratory diseases.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/trends , Lung Diseases/therapy , Humans , Lung Diseases/genetics , Mutation , Transcription Activator-Like Effector Nucleases/therapeutic use , Zinc Finger Nucleases/therapeutic use
8.
Am J Respir Cell Mol Biol ; 59(5): 635-647, 2018 11.
Article in English | MEDLINE | ID: mdl-29958015

ABSTRACT

Studies showed that TRIM72 is essential for repair of alveolar cell membrane disruptions, and exogenous recombinant human TRIM72 protein (rhT72) demonstrated tissue-mending properties in animal models of tissue injury. Here we examine the mechanisms of rhT72-mediated lung cell protection in vitro and test the efficacy of inhaled rhT72 in reducing tissue pathology in a mouse model of ventilator-induced lung injury. In vitro lung cell injury was induced by glass beads and stretching. Ventilator-induced lung injury was modeled by injurious ventilation at 30 ml/kg tidal volume. Affinity-purified rhT72 or control proteins were added into culture medium or applied through nebulization. Cellular uptake and in vivo distribution of rhT72 were detected by imaging and immunostaining. Exogenous rhT72 maintains membrane integrity of alveolar epithelial cells subjected to glass bead injury in a dose-dependent manner. Inhaled rhT72 decreases the number of fatally injured alveolar cells, and ameliorates tissue-damaging indicators and cell injury markers after injurious ventilation. Using in vitro stretching assays, we reveal that rhT72 improves both cellular resilience to membrane wounding and membrane repair after injury. Image analysis detected rhT72 uptake by rat alveolar epithelial cells, which can be inhibited by a cholesterol-disrupting agent. In addition, inhaled rhT72 distributes to the distal lungs, where it colocalizes with phosphatidylserine detection on nonpermeabilized lung slices to label wounded cells. In conclusion, our study showed that inhaled rhT72 accumulates in injured lungs and protects lung tissue from ventilator injury, the mechanisms of which include improving cell resilience to membrane wounding, localizing to injured membrane, and augmenting membrane repair.


Subject(s)
Carrier Proteins/administration & dosage , Pulmonary Alveoli/metabolism , Recombinant Proteins/administration & dosage , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/prevention & control , Wound Healing , Administration, Inhalation , Animals , Cell Membrane/metabolism , Cells, Cultured , Humans , Membrane Proteins , Mice , Pulmonary Alveoli/injuries , Pulmonary Alveoli/pathology , Rats , Ventilator-Induced Lung Injury/etiology , Ventilator-Induced Lung Injury/pathology
9.
J Cell Biochem ; 119(4): 3519-3527, 2018 04.
Article in English | MEDLINE | ID: mdl-29144017

ABSTRACT

It was found that the expression level of miR-147a was significantly increased and the pathway of PI3K/AKT was dramatically inhibited after radiation. In view of the relationship between miRNA and target genes, we put forward the question, what is the relationship between PI3K/AKT and miR-147a? In order to find the answer to the question, we used bioinformatics techniques to analyze the relationship between miR-147 (a or b) and PI3K/AKT signaling pathway. miR-147a overexpression plasmid and PDPK1 3'UTR luciferase reporter gene plasmid were constructed. Dual luciferase reporter gene system validation experiments were carried out on miR-147a and PDPK1 relationship. The verification experiments were also carried out. Bioinformatics analysis showed that there is a miR-147a binding site in the non-coding region (3'UTR) of PDPK1. In the experimental groups transfected with wild type PDPK1 gene of 3'UTR plasmid, the luciferase activity decreased (or increased) significantly in miR-147a (or inhibitor) group compared with miR-NC (or anti-miR-NC); There was no significant difference between the miR-147a group (or inhibitor) and the miR-NC group (or anti-miR-NC) in the transfection of PDPK1-3'UTR-Mut gene vector. PDPK1 was a target gene for direct regulation of miR-147a downstream. Verifying test results showed that the expression of PDPK1 mRNA and protein was reduced after overexpression of miR-147a, which was up-regulated after silencing miR-147a in TC, and V79 cells. These results suggest that miR-147a could be involved in the regulation of PDPK1 transcription by binding to the target site in PDPK1 mRNA 3'UTR, and then regulated AKT.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/genetics , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Animals , Cells, Cultured , Computational Biology , Cricetinae , HEK293 Cells , Humans , Immunoblotting , Mice , Protein Binding/radiation effects , Signal Transduction/drug effects
10.
Apoptosis ; 23(9-10): 470-483, 2018 10.
Article in English | MEDLINE | ID: mdl-29995207

ABSTRACT

The present study evaluated the protective effect of the natural compound flavonoids of Rosa roxburghii Tratt (FRT) against γ-radiation-induced apoptosis and inflammation in mouse thymus cells in vivo and in vitro. Thymus cells and mice were exposed to 60Co γ-ray at a dose of 6 Gy. The radiation treatment induced significant cell apoptosis and inflammation. Radiation increased the expressions of cleaved caspase 3/8-10, AIF, and PARP-1, and FRT could mitigate their activation and inhibit subsequent apoptosis in the thymus both in vitro or in vivo. Irradiation increased the mRNA expression of ICAM-1/VCAM-1, IL-1α/IL-6 and TNF-α/NF-κB. Our results also indicated that FRT alleviated gene expression of some inflammatory factors such as ICAM-1/VCAM-1, TNF-α/NF-κB, but not IL-1α/IL-6. Irradiation increased the protein expression levels of ICAM-1/VCAM-1, IL-1α/IL-6 and TNF-α/NF-Κb, and our results also indicated that FRT alleviated protein level expression of certain inflammatory factors such as ICAM-1, IL-1α/IL-6, TNF-α/NF-κB, but not VCAM-1. Our results suggested that FRT enhanced radioprotection at least partially by regulating caspase 3/8-10, AIF, and PARP-1 to reduce apoptosis and by regulating ICAM-1, IL-1α/IL-6, TNF-α/NF-κB to reduce inflammation.


Subject(s)
Apoptosis/drug effects , Flavonoids/administration & dosage , Inflammation/drug therapy , Rosa/chemistry , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Caspase 3/genetics , Flavonoids/chemistry , Gamma Rays/adverse effects , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Mice , NF-kappa B/genetics , Signal Transduction/drug effects , Thymus Gland/drug effects , Thymus Gland/pathology , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/genetics
11.
Int J Mol Sci ; 19(3)2018 Mar 16.
Article in English | MEDLINE | ID: mdl-29547542

ABSTRACT

Transepithelial fluid and salt re-absorption in epithelial tissues play an important role in fluid and salt homeostasis. In absorptive epithelium, fluid and salt flux is controlled by machinery mainly composed of epithelial sodium channels (ENaC), cystic fibrosis transmembrane conductance regulator (CFTR), Na⁺/H⁺ exchanger (NHE), aquaporin, and sodium potassium adenosine triphosphatase (Na⁺/K⁺-ATPase). Dysregulation of fluid and salt transport across epithelium contributes to the pathogenesis of many diseases, such as pulmonary edema and cystic fibrosis. Intracellular and extracellular signals, i.e., hormones and protein kinases, regulate fluid and salt turnover and resolution. Increasing evidence demonstrates that transepithelial fluid transport is regulated by cyclic guanosine monophosphate-dependent protein kinase (cGK) signals. cGK2 was originally identified and cloned from intestinal specimens, the presence of which has also been confirmed in the kidney and the lung. cGK2 regulates fluid and salt through ENaC, CFTR and NHE. Deficient cGK2 regulation of transepithelial ion transport was seen in acute lung injury, and cGK2 could be a novel druggable target to restore edematous disorder in epithelial tissues.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Transcytosis , Water-Electrolyte Balance , Acute Lung Injury/metabolism , Animals , Aquaporins/metabolism , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drug Discovery , Epithelial Sodium Channels/metabolism , Epithelium/metabolism , Humans , Ion Transport , Mice , Sodium-Potassium-Exchanging ATPase/metabolism
12.
Int J Mol Sci ; 19(9)2018 Aug 23.
Article in English | MEDLINE | ID: mdl-30142967

ABSTRACT

Calmodulin (CaM) is well known as an activator of calcium/calmodulin-dependent protein kinase II (CaMKII). Voltage-gated sodium channels (VGSCs) are basic signaling molecules in excitable cells and are crucial molecular targets for nervous system agents. However, the way in which Ca2+/CaM/CaMKII cascade modulates NaV1.1 IQ (isoleucine and glutamine) domain of VGSCs remains obscure. In this study, the binding of CaM, its mutants at calcium binding sites (CaM12, CaM34, and CaM1234), and truncated proteins (N-lobe and C-lobe) to NaV1.1 IQ domain were detected by pull-down assay. Our data showed that the binding of Ca2+/CaM to the NaV1.1 IQ was concentration-dependent. ApoCaM (Ca2+-free form of calmodulin) bound to NaV1.1 IQ domain preferentially more than Ca2+/CaM. Additionally, the C-lobe of CaM was the predominant domain involved in apoCaM binding to NaV1.1 IQ domain. By contrast, the N-lobe of CaM was predominant in the binding of Ca2+/CaM to NaV1.1 IQ domain. Moreover, CaMKII-mediated phosphorylation increased the binding of Ca2+/CaM to NaV1.1 IQ domain due to one or several phosphorylation sites in T1909, S1918, and T1934 of NaV1.1 IQ domain. This study provides novel mechanisms for the modulation of NaV1.1 by the Ca2+/CaM/CaMKII axis. For the first time, we uncover the effect of Ca2+, lobe-specificity and CaMKII on CaM binding to NaV1.1.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium/chemistry , Calmodulin/chemistry , NAV1.1 Voltage-Gated Sodium Channel/chemistry , Amino Acid Sequence , Binding Sites , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/genetics , Calmodulin/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HEK293 Cells , Humans , Kinetics , Molecular Docking Simulation , Mutation , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thermodynamics
13.
Cell Physiol Biochem ; 44(3): 1120-1132, 2017.
Article in English | MEDLINE | ID: mdl-29179210

ABSTRACT

Epithelial sodium channels (ENaC) play an important role in re-absorbing excessive luminal fluid by building up an osmotic Na+ gradient across the tight epithelium in the airway, the lung, the kidney, and the colon. The ENaC is a major pathway for retention of salt in kidney too. MicroRNAs (miRs), a group of non-coding RNAs that regulate gene expression at the post-transcriptional level, have emerged as a novel class of regulators for ENaC. Given the ENaC pathway is crucial for maintaining fluid homeostasis in the lung and the kidney and other cavities, we summarized the cross-talk between ENaC and miRs and recapitulated the underlying regulatory factors, including aldosterone, transforming growth factor-ß1, and vascular endothelial growth factor-A in the lung and other epithelial tissues/organs. We have compared the profiling of miRs between normal and injured mice and human lungs, which showed a significant alteration in numerous miRs in mouse models of LPS and ventilator induced ARDS. In addition, we reiterated the potential regulation of the ENaC by miRs in stem/ progenitor cell-based re-epithelialization, and identified a promising pharmaceutic target of ENaC for removing edema fluid in ARDS by mesenchymal stem cells-released paracrine. In conclusion, it seems that the interactions between miRs and scnn1s/ENaCs are critical for lung development, epithelial cell turnover in adult lungs, and re-epithelialization for repair.


Subject(s)
Epithelial Sodium Channels/metabolism , Epithelium/physiology , Lung/physiology , MicroRNAs/metabolism , Animals , Epithelium/growth & development , Humans , Regeneration , Stem Cells/cytology , Stem Cells/metabolism
15.
J Biol Chem ; 290(9): 5241-55, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25555911

ABSTRACT

Plasminogen activator inhibitor 1 (PAI-1) level is extremely elevated in the edematous fluid of acutely injured lungs and pleurae. Elevated PAI-1 specifically inactivates pulmonary urokinase-type (uPA) and tissue-type plasminogen activators (tPA). We hypothesized that plasminogen activation and fibrinolysis may alter epithelial sodium channel (ENaC) activity, a key player in clearing edematous fluid. Two-chain urokinase (tcuPA) has been found to strongly stimulate heterologous human αßγ ENaC activity in a dose- and time-dependent manner. This activity of tcuPA was completely ablated by PAI-1. Furthermore, a mutation (S195A) of the active site of the enzyme also prevented ENaC activation. By comparison, three truncation mutants of the amino-terminal fragment of tcuPA still activated ENaC. uPA enzymatic activity was positively correlated with ENaC current amplitude prior to reaching the maximal level. In sharp contrast to uPA, neither single-chain tPA nor derivatives, including two-chain tPA and tenecteplase, affected ENaC activity. Furthermore, γ but not α subunit of ENaC was proteolytically cleaved at ((177)GR↓KR(180)) by tcuPA. In summary, the underlying mechanisms of urokinase-mediated activation of ENaC include release of self-inhibition, proteolysis of γ ENaC, incremental increase in opening rate, and activation of closed (electrically "silent") channels. This study for the first time demonstrates multifaceted mechanisms for uPA-mediated up-regulation of ENaC, which form the cellular and molecular rationale for the beneficial effects of urokinase in mitigating mortal pulmonary edema and pleural effusions.


Subject(s)
Catalytic Domain , Epithelial Sodium Channels/chemistry , Protein Structure, Tertiary , Urokinase-Type Plasminogen Activator/chemistry , Animals , Binding Sites/genetics , Blotting, Western , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Female , Humans , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Kinetics , Models, Molecular , Mutation , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding , Proteolysis , Sodium/metabolism , Up-Regulation , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Xenopus laevis
16.
J Biol Chem ; 290(7): 4248-59, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25527505

ABSTRACT

The interferon-induced transmembrane proteins (IFITMs) broadly inhibit virus infections, particularly at the viral entry level. However, despite this shared ability to inhibit fusion, IFITMs differ in the potency and breadth of viruses restricted, an anomaly that is not fully understood. Here, we show that differences in the range of viruses restricted by IFITM1 are regulated by a C-terminal non-canonical dibasic sorting signal KRXX that suppresses restriction of some viruses by governing its intracellular distribution. Replacing the two basic residues with alanine (KR/AA) increased restriction of jaagsiekte sheep retrovirus and 10A1 amphotropic murine leukemia virus. Deconvolution microscopy revealed an altered subcellular distribution for KR/AA, with fewer molecules in LAMP1-positive lysosomes balanced by increased levels in CD63-positive multivesicular bodies, where jaagsiekte sheep retrovirus pseudovirions are colocalized. IFITM1 binds to cellular adaptor protein complex 3 (AP-3), an association that is lost when the dibasic motif is altered. Although knockdown of AP-3 itself decreases some virus entry, expression of parental IFITM1, but not its KR/AA mutant, potentiates inhibition of viral infections in AP-3 knockdown cells. By using the substituted cysteine accessibility method, we provide evidence that IFITM1 adopts more than one membrane topology co-existing in cellular membranes. Because the C-terminal dibasic sorting signal is unique to human IFITM1, our results provide novel insight into understanding the species- and virus-specific antiviral effect of IFITMs.


Subject(s)
Adaptor Protein Complex 3/metabolism , Antigens, Differentiation/metabolism , Cell Membrane/metabolism , Jaagsiekte sheep retrovirus/physiology , Protein Sorting Signals/physiology , Virus Internalization , Animals , Antigens, Differentiation/genetics , Blotting, Western , Cell Fusion , Cells, Cultured , Humans , Immunoprecipitation , Lysosomes/metabolism , Mutation/genetics , Protein Transport , Sheep , Virus Diseases/virology , Virus Replication
18.
Am J Physiol Lung Cell Mol Physiol ; 307(8): L609-17, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25172911

ABSTRACT

Epithelial sodium channels (ENaC) govern transepithelial salt and fluid homeostasis. ENaC contributes to polarization, apoptosis, epithelial-mesenchymal transformation, etc. Fibrinolytic proteases play a crucial role in virtually all of these processes and are elaborated by the airway epithelium. We hypothesized that urokinase-like plasminogen activator (uPA) regulates ENaC function in airway epithelial cells and tested that possibility in primary murine tracheal epithelial cells (MTE). Both basal and cAMP-activated Na(+) flow through ENaC were significantly reduced in monolayers of uPA-deficient cells. The reduction in ENaC activity was further confirmed in basolateral membrane-permeabilized cells. A decrease in the Na(+)-K(+)-ATPase activity in the basolateral membrane could contribute to the attenuation of ENaC function in intact monolayer cells. Dysfunctional fluid resolution was seen in uPA-disrupted cells. Administration of uPA and plasmin partially restores ENaC activity and fluid reabsorption by MTEs. ERK1/2, but not Akt, phosphorylation was observed in the cells and lungs of uPA-deficient mice. On the other hand, cleavage of γ ENaC is significantly depressed in the lungs of uPA knockout mice vs. those of wild-type controls. Expression of caspase 8, however, did not differ between wild-type and uPA(-/-) mice. In addition, uPA deficiency did not alter transepithelial resistance. Taken together, the mechanisms for the regulation of ENaC by uPA in MTEs include augmentation of Na(+)-K(+)-ATPase, proteolysis, and restriction of ERK1/2 phosphorylation. We demonstrate for the first time that ENaC may serve as a downstream signaling target by which uPA controls the biophysical profiles of airway fluid and epithelial function.


Subject(s)
Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Trachea/metabolism , Urokinase-Type Plasminogen Activator/physiology , Animals , Apoptosis , Cell Membrane Permeability , Cells, Cultured , Epithelial Cells/cytology , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/genetics , Immunoblotting , Ion Transport , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oocytes/metabolism , Phosphorylation , Proteolysis , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , RNA, Small Interfering/genetics , Trachea/cytology , Xenopus laevis/metabolism
19.
Am J Pathol ; 183(1): 131-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23665346

ABSTRACT

Alveolar type II epithelial cell (ATII) apoptosis and proliferation of mesenchymal cells are the hallmarks of idiopathic pulmonary fibrosis, a devastating disease of unknown cause characterized by alveolar epithelial injury and progressive fibrosis. We used a mouse model of bleomycin (BLM)-induced lung injury to understand the involvement of p53-mediated changes in urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor-1 (PAI-1) levels in the regulation of alveolar epithelial injury. We found marked induction of p53 in ATII cells from mice exposed to BLM. Transgenic mice expressing transcriptionally inactive dominant negative p53 in ATII cells showed augmented apoptosis, whereas those deficient in p53 resisted BLM-induced ATII cell apoptosis. Inhibition of p53 transcription failed to suppress PAI-1 or induce uPA mRNA in BLM-treated ATII cells. ATII cells from mice with BLM injury showed augmented binding of p53 to uPA, uPA receptor (uPAR), and PAI-1 mRNA. p53-binding sequences from uPA, uPAR, and PAI-1 mRNA 3' untranslated regions neither interfered with p53 DNA binding activity nor p53-mediated promoter transactivation. However, increased expression of p53-binding sequences from uPA, uPAR, and PAI-1 mRNA 3' untranslated regions in ATII cells suppressed PAI-1 and induced uPA after BLM treatment, leading to inhibition of ATII cell apoptosis and pulmonary fibrosis. Our findings indicate that disruption of p53-fibrinolytic system cross talk may serve as a novel intervention strategy to prevent lung injury and pulmonary fibrosis.


Subject(s)
Lung Injury/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/metabolism , Tumor Suppressor Protein p53/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Biomarkers/metabolism , Bleomycin , Blotting, Western , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Immunohistochemistry , Lung Injury/chemically induced , Lung Injury/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology
20.
BMC Cancer ; 14: 544, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25069990

ABSTRACT

BACKGROUND: Ectopic TSH-secreting pituitary adenoma (TSH-oma) is a very unusual disorder. To date, there are only four cases reported. It is difficult to distinguish ectopic cases from both regular TSH-omas and resistance to thyroid hormone (RTH). CASE PRESENTATION: A newly identified case of ectopic TSH-oma arising from the nasal pharynx was described, and reports of four prior cases were reviewed. The patient was a 41-year-old male who developed what appeared to be typical hyperthyroidism and atrial fibrillation in 2009. Thyroid function tests showed elevated basal levels of free T3 (FT3, 24.08 pmol/L), free T4 (FT4, 75.73 pmol/L), and serum TSH (7.26 µIU/ml). Both TSH-oma and resistance to thyroid hormone syndrome were considered. TRH stimulating test was negative, whereas octreotide inhibition test showed a reduction in TSH by 30.8%. Furthermore, a large space-occupying lesion located at the nasopharynx was found by computed tomography and magnetic resonance imaging (MRI). A normal pituitary was visualized. Ectopic TSH-oma was preliminarily established. Using an endoscopic endonasal approach, the tumor was resected. Histological features and immunophenotypes were consistent with those of TSH-secreting tumor. The levels of both free thyroxine and TSH returned to normal ranges the day after surgery and remained within normal range for 48 months. CONCLUSIONS: Although exceedingly rare, ectopic TSH-oma should be considered for patients with inappropriate secretion of TSH with hyperthyroidism and pituitary tumor undetectable by computed tomography and MRI. To our knowledge, this is the first case followed up more than 4 years. The characteristics and successful interventions summarized in this report provide a guideline for clinicians.


Subject(s)
Adenoma/pathology , Pituitary Neoplasms/pathology , Thyrotropin/metabolism , Adenoma/metabolism , Adenoma/surgery , Adult , Humans , Hyperthyroidism/metabolism , Hyperthyroidism/pathology , Male , Octreotide/pharmacology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/surgery , Rare Diseases/metabolism , Rare Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL