Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 699
Filter
1.
Elife ; 102021 09 29.
Article in English | MEDLINE | ID: mdl-34586063

ABSTRACT

Lung epithelial progenitors differentiate into alveolar type 1 (AT1) and type 2 (AT2) cells. These cells form the air-blood interface and secrete surfactant, respectively, and are essential for lung maturation and function. Current protocols to derive and culture alveolar cells do not faithfully recapitulate the architecture of the distal lung, which influences cell fate patterns in vivo. Here, we report serum-free conditions that allow for growth and differentiation of mouse distal lung epithelial progenitors. We find that Collagen I promotes the differentiation of flattened, polarized AT1 cells. Using these organoids, we performed a chemical screen to investigate WNT signaling in epithelial differentiation. We identify an association between Casein Kinase activity and maintenance of an AT2 expression signature; Casein Kinase inhibition leads to an increase in AT1/progenitor cell ratio. These organoids provide a simplified model of alveolar differentiation and constitute a scalable screening platform to identify and analyze cell differentiation mechanisms.


Subject(s)
Cell Differentiation , Pulmonary Alveoli/cytology , Stem Cells/cytology , Animals , Casein Kinases/antagonists & inhibitors , Casein Kinases/metabolism , Cells, Cultured , Collagen Type I/metabolism , Culture Media, Serum-Free , Epithelial Cells/cytology , Epithelial Cells/metabolism , Genetic Markers , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/embryology , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/metabolism , Transcription, Genetic , Wnt Signaling Pathway
2.
Angiogenesis ; 24(3): 677-693, 2021 08.
Article in English | MEDLINE | ID: mdl-33770321

ABSTRACT

Endothelial barrier disruption and vascular leak importantly contribute to organ dysfunction and mortality during inflammatory conditions like sepsis and acute respiratory distress syndrome. We identified the kinase Arg/Abl2 as a mediator of endothelial barrier disruption, but the role of Arg in endothelial monolayer regulation and its relevance in vivo remain poorly understood. Here we show that depletion of Arg in endothelial cells results in the activation of both RhoA and Rac1, increased cell spreading and elongation, redistribution of integrin-dependent cell-matrix adhesions to the cell periphery, and improved adhesion to the extracellular matrix. We further show that Arg is activated in the endothelium during inflammation, both in murine lungs exposed to barrier-disruptive agents, and in pulmonary microvessels of septic patients. Importantly, Arg-depleted endothelial cells were less sensitive to barrier-disruptive agents. Despite the formation of F-actin stress fibers and myosin light chain phosphorylation, Arg depletion diminished adherens junction disruption and intercellular gap formation, by reducing the disassembly of cell-matrix adhesions and cell retraction. In vivo, genetic deletion of Arg diminished vascular leak in the skin and lungs, in the presence of a normal immune response. Together, our data indicate that Arg is a central and non-redundant regulator of endothelial barrier integrity, which contributes to cell retraction and gap formation by increasing the dynamics of adherens junctions and cell-matrix adhesions in a Rho GTPase-dependent fashion. Therapeutic inhibition of Arg may provide a suitable strategy for the treatment of a variety of clinical conditions characterized by vascular leak.


Subject(s)
Extracellular Matrix/metabolism , Gap Junctions/enzymology , Human Umbilical Vein Endothelial Cells/enzymology , Protein-Tyrosine Kinases/metabolism , Pulmonary Alveoli/enzymology , Animals , Cell Adhesion/genetics , Enzyme Activation , Extracellular Matrix/genetics , Gap Junctions/genetics , Humans , Inflammation/enzymology , Inflammation/genetics , Mice , Mice, Knockout , Protein-Tyrosine Kinases/genetics
3.
Part Fibre Toxicol ; 18(1): 11, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33706759

ABSTRACT

BACKGROUND: Air pollution exposure and idiopathic pulmonary fibrosis (IPF) cause a poor prognosis after SARS-CoV-2 infection, but the underlying mechanisms are not well explored. Angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) are the keys to the entry of SARS-CoV-2. We therefore hypothesized that air pollution exposure and IPF may increase the expression of ACE2 and TMPRSS2 in the lung alveolar region. We measured their expression levels in lung tissues of control non-IPF and IPF patients, and used murine animal models to study the deterioration of IPF caused by particulate matter (PM) and the molecular pathways involved in the expression of ACE2 and TMPRSS2. RESULTS: In non-IPF patients, cells expressing ACE2 and TMPRSS2 were limited to human alveolar cells. ACE2 and TMPRSS2 were largely upregulated in IPF patients, and were co-expressed by fibroblast specific protein 1 (FSP-1) + lung fibroblasts in human pulmonary fibrotic tissue. In animal models, PM exposure increased the severity of bleomycin-induced pulmonary fibrosis. ACE2 and TMPRSS2 were also expressed in FSP-1+ lung fibroblasts in bleomycin-induced pulmonary fibrosis, and when combined with PM exposure, they were further upregulated. The severity of pulmonary fibrosis and the expression of ACE2 and TMPRSS2 caused by PM exposure were blocked by deletion of KC, a murine homologue of IL-8, or treatment with reparixin, an inhibitor of IL-8 receptors CXCR1/2. CONCLUSIONS: These data suggested that risk of SARS-CoV-2 infection and COVID-19 disease severity increased by air pollution exposure and underlying IPF. It can be mediated through upregulating ACE2 and TMPRSS2 in pulmonary fibroblasts, and prevented by blocking the IL-8/CXCR1/2 pathway.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/etiology , Idiopathic Pulmonary Fibrosis/complications , Particulate Matter/toxicity , SARS-CoV-2 , Serine Endopeptidases/genetics , Angiotensin-Converting Enzyme 2/physiology , Animals , Humans , Interleukin-8/physiology , Male , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/enzymology , Serine Endopeptidases/physiology , Up-Regulation
4.
Lung ; 197(2): 227-233, 2019 04.
Article in English | MEDLINE | ID: mdl-30759273

ABSTRACT

BACKGROUND: Pancreatic digestive enzymes present in meconium might be responsible for meconium-induced lung injury. The local Renin Angiotensin System plays an important role in lung injury and inflammation. Particularly, angiotensin converting enzyme-2 (ACE-2) has been identified as a protective lung enzyme against the insult. ACE-2 converts pro-apoptotic Angiotensin II to anti-apoptotic Angiotensin 1-7. However, the effect of meconium on ACE-2 has never been studied before. OBJECTIVE: To study the effect of meconium on ACE-2, and whether inhibition of proteolytic enzymes present in the meconium reverses its effects on ACE-2. METHODS: Alveolar epithelial A549 cells were exposed to F-12 medium, 2.5% meconium, meconium + a protease inhibitor cocktail (PIc) and PIc alone for 16 h. At the end of incubation, apoptosis was measured with a nuclear fragmentation assay and cell lysates were collected for ACE-2 immunoblotting and enzyme activity. RESULTS: Meconium caused a fourfold increase in apoptotic nuclei (p < 0.001). The pro-apoptotic effect of meconium can be reversed by PIc. Meconium reduced ACE-2 enzyme activity by cleaving ACE-2 into a fragment detected at ~ 37 kDa by immunoblot. PIc prevented the degradation of ACE-2 and restored 50% of ACE-2 activity (p < 0.05). CONCLUSION: These data suggest that meconium causes degradation of lung protective ACE-2 by proteolytic enzymes present in meconium, since the effects of meconium can be reversed by PIc.


Subject(s)
Epithelial Cells/enzymology , Meconium Aspiration Syndrome/enzymology , Meconium/enzymology , Peptide Hydrolases/metabolism , Peptidyl-Dipeptidase A/metabolism , Pulmonary Alveoli/enzymology , A549 Cells , Angiotensin-Converting Enzyme 2 , Apoptosis , Enzyme Stability , Epithelial Cells/pathology , Humans , Meconium Aspiration Syndrome/pathology , Proteolysis , Pulmonary Alveoli/pathology
5.
Int J Rheum Dis ; 22(4): 686-694, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30666825

ABSTRACT

AIM: To investigate the potential therapeutic efficacy of iguratimod (IGU) on bleomycin (BLM)-induced pulmonary fibrosis in mice. METHODS: A total of 75 C57BL/6 mice were randomly and evenly divided into control group, BLM (5 mg/kg) group, BLM + IGU (90 mg/kg) group, BLM + methylprednisolone (MP, 10 mg/kg) group and BLM + pirfenidone (PF, 100 mg/kg) group. The mice were sacrificed on day 7, 14 and 28. The lung tissue was examined by hematoxylin and eosin staining and Masson staining to evaluate the degree of alveolitis and fibrosis, and serum cytokines were measured. RESULTS: Histopathological results showed that IGU attenuated BLM-induced alveolar inflammation and decreased collagen deposition in lung tissue from day 7 till day 28. Both the pathological alveolitis and fibrosis scores in the drug-treated groups (IGU group, MP group and PF group) were decreased dramatically compared with the BLM group on day 7, 14 and 28 (P < 0.05). There were no statistical significances among these three groups. Cytokine profile showed that IGU decreased the level of tumor necrosis factor-α (TNF-α), interleukin (IL)-1, IL-6 and matrix metalloproteinase (MMP)-9 which were up-regulated by BLM on day 7, 14 and 28 (P < 0.05). Furthermore, there is a strong correlation between the severity of pulmonary fibrosis and serum MMP-9 levels. CONCLUSION: IGU can decrease BLM-induced pulmonary fibrosis, and the anti-fibrotic effect of IGU is mediated partly via inhibition of MMP-9, which suggests that IGU could potentially be an effective therapeutic strategy for pulmonary fibrosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bleomycin , Chromones/pharmacology , Collagen/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Pneumonia/prevention & control , Pulmonary Alveoli/drug effects , Pulmonary Fibrosis/prevention & control , Sulfonamides/pharmacology , Airway Remodeling/drug effects , Animals , Cytokines/blood , Disease Models, Animal , Inflammation Mediators/blood , Male , Mice, Inbred C57BL , Pneumonia/chemically induced , Pneumonia/enzymology , Pneumonia/pathology , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology
6.
FEBS J ; 285(16): 3056-3076, 2018 08.
Article in English | MEDLINE | ID: mdl-29935061

ABSTRACT

The generation, maturation and remodelling of the extracellular matrix (ECM) are essential for the formation of alveoli during lung development. Alveoli formation is disturbed in preterm infants that develop bronchopulmonary dysplasia (BPD), where collagen fibres are malformed, and perturbations to lung ECM structures may underlie BPD pathogenesis. Malformed ECM structures might result from abnormal protein cross-linking, in part attributable to the increased expression and activity of transglutaminase 2 (TGM2) that have been noted in affected patient lungs, as well as in hyperoxia-based BPD animal models. The objective of the present study was to assess whether TGM2 plays a causal role in normal and aberrant lung alveolarization. Targeted deletion of Tgm2 in C57BL/6J mice increased septal thickness and reduced gas-exchange surface area in otherwise normally developing lungs. During aberrant lung alveolarization that occurred under hyperoxic conditions, collagen structures in Tgm2-/- mice were partially protected from the impact of hyperoxia, where normal dihydroxylysinonorleucine and hydroxylysylpiridinoline collagen cross-link abundance was restored; however, the lung alveolar architecture remained abnormal. Inhibition of transglutaminases (including TGM2) with cysteamine appreciably reduced transglutaminase activity in vivo, as assessed by Nε -(γ-l-glutamyl)-l-lysine abundance and TGM catalytic activity, and restored normal dihydroxylysinonorleucine and hydroxylysylpiridinoline collagen cross-link abundance under pathological conditions. Furthermore, a moderate improvement in alveoli size and gas-exchange surface density was noted in cysteamine-treated mouse lungs in which BPD was modelled. These data indicate that TGM2 plays a role in normal lung alveolarization, and contributes to the formation of aberrant ECM structures during disordered lung alveolarization.


Subject(s)
Bronchopulmonary Dysplasia/enzymology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Pulmonary Alveoli/enzymology , Transglutaminases/genetics , Transglutaminases/metabolism , Animals , Bronchopulmonary Dysplasia/genetics , Collagen/metabolism , Collagen/ultrastructure , Cysteamine/pharmacology , Dipeptides/immunology , Dipeptides/metabolism , Disease Models, Animal , Extracellular Matrix/enzymology , Extracellular Matrix/pathology , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Hyperoxia/genetics , Lung/drug effects , Lung/enzymology , Lung/growth & development , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Protein Glutamine gamma Glutamyltransferase 2 , Pulmonary Alveoli/pathology , Pulmonary Alveoli/ultrastructure
7.
Prog Mol Biol Transl Sci ; 148: 1-29, 2017.
Article in English | MEDLINE | ID: mdl-28662821

ABSTRACT

Normal gas exchange after birth requires functional lung alveolar units that are lined with epithelial cells, parts of which are intricately fused with microvascular capillaries. A significant phase of alveolar lung development occurs in the perinatal period, continues throughout early stages in life, and requires activation of matrix-remodeling enzymes. Failure to achieve an optimum number of alveoli during lung maturation can cause several untoward medical consequences including disabling obstructive and/or restrictive lung diseases that limit physiological endurance and increase mortality. Several members of the matrix metalloproteinase (MMP) family are critical in lung remodeling before and after birth; however, their resurgence in response to environmental factors, infection, and injury can also compromise lung function. Therefore, temporal expression, regulation, and function of MMPs play key roles in developing and maintaining adequate oxygenation under steady state, as well as in diseased conditions. Broadly, with the exception of MMP2 and MMP14, most deletional mutations of MMPs fail to perturb lung development; however, their individual absence can alter the pathophysiology of respiratory diseases. Specifically, under stressed conditions such as acute respiratory infection and allergic inflammation, MMP2 and MMP9 can play a protective role through bacterial clearance and production of chemotactic gradient, while loss of MMP12 can protect mice from smoke-induced lung disease. Therefore, better understanding of the expression and function of MMPs under normal lung development and their resurgence in response respiratory diseases could provide new therapeutic options in the future.


Subject(s)
Lung/enzymology , Lung/pathology , Matrix Metalloproteinases/metabolism , Wound Healing , Animals , Humans , Lung/embryology , Lung Injury/enzymology , Lung Injury/pathology , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/pathology
8.
Toxicol Sci ; 157(1): 260-271, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28201809

ABSTRACT

Prolonged hyperoxia contributes to bronchopulmonary dysplasia (BPD) in preterm infants. ß-Naphthoflavone (BNF) is a potent inducer of cytochrome P450 (CYP)1A enzymes, which have been implicated in hyperoxic injuries in adult mice. In this investigation, we tested the hypothesis that newborn mice lacking the Cyp1a1 gene would be more susceptible to hyperoxic lung injury than wild-type (WT) mice and that postnatal BNF treatment would rescue this phenotype by mechanisms involving CYP1A and/or NAD(P)H quinone oxidoreductase (NQO1) enzymes. Newborn WT or Cyp1a1-null mice were treated with BNF (10 mg/kg) or the vehicle corn oil (CO) i.p., from postnatal day (PND) 2 to 14 once every other day, while being maintained in room air or hyperoxia (85% O2) for 14 days. Both genotypes showed lung injury, inflammation, and alveolar simplification in hyperoxia, with Cyp1a1-null mice displaying increased susceptibility compared to WT mice. BNF treatment resulted in significant attenuation of lung injury and inflammation, with improved alveolarization in both WT and Cyp1a1-null mice. BNF exposed normoxic or hyperoxic WT mice showed increased expression of hepatic CYP1A1/1A2, pulmonary CYP1A1, and NQO1 expression at both mRNA and protein levels, compared with vehicle controls. However, BNF caused greater induction of hepatic CYP1A2 and pulmonary NQO1 enzymes in the Cyp1a1-null mice, suggesting that BNF protects against hyperoxic lung injury in WT and Cyp1a1-null mice through the induction of CYP1A and NQO1 enzymes. Further studies on the protective role of flavonoids against hyperoxic lung injury in newborns could lead to novel strategies for the prevention and/or treatment of BPD.


Subject(s)
Bronchopulmonary Dysplasia/etiology , Cytochrome P-450 CYP1A1/genetics , Infant, Premature , Oxygen/administration & dosage , beta-Naphthoflavone/administration & dosage , Animals , Animals, Newborn , Blotting, Western , Bronchopulmonary Dysplasia/genetics , Genetic Predisposition to Disease , Humans , Infant, Newborn , Mice , Mice, Knockout , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/enzymology
9.
Free Radic Biol Med ; 101: 482-490, 2016 12.
Article in English | MEDLINE | ID: mdl-27840320

ABSTRACT

RATIONALE: Alveolar epithelial cell (AEC) injury and mitochondrial dysfunction are important in the development of lung fibrosis. Our group has shown that in the asbestos exposed lung, the generation of mitochondrial reactive oxygen species (ROS) in AEC mediate mitochondrial DNA (mtDNA) damage and apoptosis which are necessary for lung fibrosis. These data suggest that mitochondrial-targeted antioxidants should ameliorate asbestos-induced lung. OBJECTIVE: To determine whether transgenic mice that express mitochondrial-targeted catalase (MCAT) have reduced lung fibrosis following exposure to asbestos or bleomycin and, if so, whether this occurs in association with reduced AEC mtDNA damage and apoptosis. METHODS: Crocidolite asbestos (100µg/50µL), TiO2 (negative control), bleomycin (0.025 units/50µL), or PBS was instilled intratracheally in 8-10 week-old wild-type (WT - C57Bl/6J) or MCAT mice. The lungs were harvested at 21d. Lung fibrosis was quantified by collagen levels (Sircol) and lung fibrosis scores. AEC apoptosis was assessed by cleaved caspase-3 (CC-3)/Surfactant protein C (SFTPC) immunohistochemistry (IHC) and semi-quantitative analysis. AEC (primary AT2 cells from WT and MCAT mice and MLE-12 cells) mtDNA damage was assessed by a quantitative PCR-based assay, apoptosis was assessed by DNA fragmentation, and ROS production was assessed by a Mito-Sox assay. RESULTS: Compared to WT, crocidolite-exposed MCAT mice exhibit reduced pulmonary fibrosis as measured by lung collagen levels and lung fibrosis score. The protective effects in MCAT mice were accompanied by reduced AEC mtDNA damage and apoptosis. Similar findings were noted following bleomycin exposure. Euk-134, a mitochondrial SOD/catalase mimetic, attenuated MLE-12 cell DNA damage and apoptosis. Finally, compared to WT, asbestos-induced MCAT AT2 cell ROS production was reduced. CONCLUSIONS: Our finding that MCAT mice have reduced pulmonary fibrosis, AEC mtDNA damage and apoptosis following exposure to asbestos or bleomycin suggests an important role for AEC mitochondrial H2O2-induced mtDNA damage in promoting lung fibrosis. We reason that strategies aimed at limiting AEC mtDNA damage arising from excess mitochondrial H2O2 production may be a novel therapeutic target for mitigating pulmonary fibrosis.


Subject(s)
Catalase/genetics , DNA, Mitochondrial/drug effects , Epithelial Cells/drug effects , Mitochondria/drug effects , Mitochondrial Proteins/genetics , Pulmonary Alveoli/drug effects , Pulmonary Fibrosis/prevention & control , Administration, Inhalation , Animals , Asbestos , Bleomycin , Caspase 3/genetics , Caspase 3/metabolism , Catalase/metabolism , Collagen/antagonists & inhibitors , Collagen/genetics , Collagen/metabolism , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/metabolism , Epithelial Cells/enzymology , Epithelial Cells/pathology , Gene Expression , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins , Intubation, Intratracheal , Mice , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Organometallic Compounds/pharmacology , Peptides/genetics , Peptides/metabolism , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Pulmonary Surfactant-Associated Protein C , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Salicylates/pharmacology , Transgenes
10.
Am J Respir Cell Mol Biol ; 55(6): 767-778, 2016 12.
Article in English | MEDLINE | ID: mdl-27438994

ABSTRACT

In premature infants, sepsis is associated with alveolar simplification manifesting as bronchopulmonary dysplasia. The redox-dependent mechanisms underlying sepsis-induced inflammation and alveolar remodeling in the immature lung remain unclear. We developed a neonatal mouse model of sepsis-induced lung injury to investigate whether nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) regulates Toll-like receptor (TLR)-mediated inflammation and alveolar remodeling. Six-day-old NOX2+/+ and NOX2-/- mice were injected with intraperitoneal LPS to induce sepsis. Lung inflammation and canonical TLR signaling were assessed 24 hours after LPS. Alveolar development was examined in 15-day-old mice after LPS on Day 6. The in vivo efficacy of a NOX2 inhibitor (NOX2-I) on NOX2 complex assembly and sepsis-induced lung inflammation were examined. Lung cytokine expression and neutrophil influx induced with sepsis in NOX2+/+ mice was decreased by >50% in NOX2-/- mice. LPS-induced TLR4 signaling evident by inhibitor of NF-κB kinase-ß and mitogen-activated protein kinase phosphorylation, and nuclear factor-κB/AP-1 translocation were attenuated in NOX2-/- mice. LPS increased matrix metalloproteinase 9 while decreasing elastin and keratinocyte growth factor levels in NOX2+/+ mice. An LPS-induced increase in matrix metalloproteinase 9 and decrease in fibroblast growth factor 7 and elastin were not evident in NOX2-/- mice. An LPS-induced reduction in radial alveolar counts and increased mean linear intercepts were attenuated in NOX2-/- mice. LPS-induced NOX2 assembly evident by p67phox/gp91phox coimmunoprecipitation was disrupted with NOX2-I. NOX2-I also mitigated LPS-induced cytokine expression, TLR pathway signaling, and alveolar simplification. In a mouse model of neonatal sepsis, NOX2 regulates proinflammatory TLR signaling and alveolar remodeling induced by a single dose of LPS. Our results provide mechanistic insight into the regulation of sepsis-induced alveolar remodeling in the developing lung.


Subject(s)
Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Pneumonia/enzymology , Pneumonia/pathology , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/growth & development , Acute Disease , Animals , Biomarkers/metabolism , Cytokines/metabolism , Extracellular Matrix/metabolism , Lipopolysaccharides , Membrane Glycoproteins/deficiency , Mice , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NF-kappa B/metabolism , Pneumonia/metabolism , Pulmonary Alveoli/pathology , Sepsis/complications , Sepsis/metabolism , Sepsis/pathology , Toll-Like Receptors/metabolism , Transcription Factor AP-1/metabolism
11.
Respir Res ; 17(1): 71, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27301375

ABSTRACT

BACKGROUND: Gastric contents aspiration in humans is a risk factor for severe respiratory failure with elevated mortality. Although aspiration-induced local lung inflammation has been studied in animal models, little is known about extrapulmonary effects of aspiration. We investigated whether a single orotracheal instillation of whole gastric fluid elicits a liver acute phase response and if this response contributes to enrich the alveolar spaces with proteins having antiprotease activity. METHODS: In anesthetized Sprague-Dawley rats receiving whole gastric fluid, we studied at different times after instillation (4 h -7 days): changes in blood cytokines and acute phase proteins (fibrinogen and the antiproteases alpha1-antitrypsin and alpha2-macroglobulin) as well as liver mRNA expression of the two antiproteases. The impact of the systemic changes on lung antiprotease defense was evaluated by measuring levels and bioactivity of antiproteases in broncho-alveolar lavage fluid (BALF). Markers of alveolar-capillary barrier derangement were also studied. Non-parametric ANOVA (Kruskall-Wallis) and linear regression analysis were used. RESULTS: Severe peribronchiolar injury involving edema, intra-alveolar proteinaceous debris, hemorrhage and PMNn cell infiltration was seen in the first 24 h and later resolved. Despite a large increase in several lung cytokines, only IL-6 was found elevated in blood, preceding increased liver expression and blood concentration of both antiproteases. These changes, with an acute phase response profile, were significantly larger for alpha2-macroglobulin (40-fold increment in expression with 12-fold elevation in blood protein concentration) than for alpha1-antitrypsin (2-3 fold increment in expression with 0.5-fold elevation in blood protein concentration). Both the increment in capillary-alveolar antiprotease concentration gradient due to increased antiprotease liver synthesis and a timely-associated derangement of the alveolar-capillary barrier induced by aspiration, contributed a 58-fold and a 190-fold increase in BALF alpha1-antitrypsin and alpha2-macroglobulin levels respectively (p < 0.001). CONCLUSIONS: Gastric contents-induced acute lung injury elicits a liver acute phase response characterized by increased mRNA expression of antiproteases and elevation of blood antiprotease concentrations. Hepatic changes act in concert with derangement of the alveolar capillary barrier to enrich alveolar spaces with antiproteases. These findings may have significant implications decreasing protease burden, limiting injury in this and other models of acute lung injury and likely, in recurrent aspiration.


Subject(s)
Acute Lung Injury/enzymology , Acute-Phase Reaction/enzymology , Liver/metabolism , Pregnancy-Associated alpha 2-Macroglobulins/biosynthesis , Pulmonary Alveoli/enzymology , Respiratory Aspiration of Gastric Contents/complications , alpha 1-Antitrypsin/biosynthesis , Acute Lung Injury/blood , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Acute-Phase Reaction/blood , Acute-Phase Reaction/etiology , Acute-Phase Reaction/pathology , Animals , Blood-Air Barrier/enzymology , Blood-Air Barrier/pathology , Disease Models, Animal , Enzyme Induction , Inflammation Mediators/blood , Interleukin-6/blood , Male , Pregnancy-Associated alpha 2-Macroglobulins/genetics , Pulmonary Alveoli/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats, Sprague-Dawley , Time Factors , alpha 1-Antitrypsin/blood , alpha 1-Antitrypsin/genetics
12.
J Biol Chem ; 290(52): 30813-29, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26518879

ABSTRACT

Mutations of human telomerase RNA component (TERC) and telomerase reverse transcriptase (TERT) are associated with a subset of lung aging diseases, but the mechanisms by which TERC and TERT participate in lung diseases remain unclear. In this report, we show that knock-out (KO) of the mouse gene Terc or Tert causes pulmonary alveolar stem cell replicative senescence, epithelial impairment, formation of alveolar sacs, and characteristic inflammatory phenotype. Deficiency in TERC or TERT causes a remarkable elevation in various proinflammatory cytokines, including IL-1, IL-6, CXCL15 (human IL-8 homolog), IL-10, TNF-α, and monocyte chemotactic protein 1 (chemokine ligand 2 (CCL2)); decrease in TGF-ß1 and TGFßRI receptor in the lungs; and spillover of IL-6 and CXCL15 into the bronchoalveolar lavage fluids. In addition to increased gene expressions of α-smooth muscle actin and collagen 1α1, suggesting myofibroblast differentiation, TERC deficiency also leads to marked cellular infiltrations of a mononuclear cell population positive for the leukocyte common antigen CD45, low-affinity Fc receptor CD16/CD32, and pattern recognition receptor CD11b in the lungs. Our data demonstrate for the first time that telomerase deficiency triggers alveolar stem cell replicative senescence-associated low-grade inflammation, thereby driving pulmonary premature aging, alveolar sac formation, and fibrotic lesion.


Subject(s)
Lung Diseases/immunology , Pulmonary Alveoli/enzymology , Stem Cells/cytology , Telomerase/deficiency , Animals , Cellular Senescence , Female , Humans , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lung/cytology , Lung/immunology , Lung Diseases/enzymology , Lung Diseases/genetics , Lung Diseases/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , RNA/genetics , Stem Cells/immunology , Telomerase/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
13.
Lipids ; 50(12): 1259-71, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26449236

ABSTRACT

The purpose of the study was to assess a fluorimetric assay for the determination of total phospholipase A(2) (PLA(2)) activity in biological samples introducing the innovation of immobilized substrates on crosslinked polymeric membranes. The immobilized C(12)-NBD-PtdCho, a fluorescent analogue of phosphatidylcholine, exhibited excellent stability for 3 months at 4 °C and was not desorbed in the aqueous reaction mixture during analysis. The limit of detection was 0.5 pmol FA (0.2 pg) and the linear part of the response curve extended from 1 up to 190 nmol FA/h/mL sample. The intra- and inter-day relative standard deviations (%RSD), were ≤6 and ≤9 %, respectively. Statistical comparison with other fluorescent methods showed excellent correlation and agreement. Semiempirical calculations showed a fair amount of electrostatic interaction between the NBD-labeled substrate and the crosslinked polyvinyl alcohol with the styryl pyridinium residues (PVA-SbQ) material, from the plane of which, the sn-2 acyl chain of the phospholipid stands out and is accessible by PLA(2). Atomic Force Microscopy revealed morphological alterations of the immobilized substrate after the reaction with PLA(2). Mass spectrometry showed that only C(12)-NBD-FA, the PLA(2 )hydrolysis product, was detected in the reaction mixture, indicating that PLA(2) recognizes PVA-SbQ/C(12)-NBD-PtdCho as a surface to perform catalysis.


Subject(s)
4-Chloro-7-nitrobenzofurazan/analogs & derivatives , Phosphatidylcholines/metabolism , Phospholipases A2/metabolism , Pulmonary Alveoli/enzymology , Respiratory Mucosa/enzymology , 4-Chloro-7-nitrobenzofurazan/chemistry , 4-Chloro-7-nitrobenzofurazan/metabolism , Animals , Chromatography, High Pressure Liquid , Computational Biology , Feasibility Studies , Fluorescent Dyes/chemistry , Humans , Hydrogen-Ion Concentration , Limit of Detection , Membranes, Artificial , Microscopy, Atomic Force , Phosphatidylcholines/chemistry , Phospholipases A2/blood , Pilot Projects , Polyvinyl Alcohol/analogs & derivatives , Polyvinyl Alcohol/chemistry , Pyridinium Compounds/chemistry , Reproducibility of Results , Spectrometry, Fluorescence , Stereoisomerism , Substrate Specificity , Sus scrofa
14.
Pediatr Res ; 78(6): 634-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26322414

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) worsens clinical outcomes in former preterm infants with bronchopulmonary dysplasia (BPD). Oxidant stress disrupts alveolar and vascular development in models of BPD. Bleomycin causes oxidative stress and induces BPD and PAH in neonatal rats. Disruption in the vascular endothelial growth factor (VEGF) and nitric oxide signaling pathways contributes to BPD. We hypothesized that loss of EC-SOD would worsen PAH associated with BPD in a neonatal mouse model of bleomycin-induced BPD by disrupting the VEGF/NO signaling pathway. METHODS: Neonatal wild-type mice (WT), and mice lacking EC-SOD (EC-SOD KO) received intraperitoneal bleomycin (2 units/kg) or phosphate-buffered saline (PBS) three times weekly and were evaluated at weeks 3 or 4. RESULTS: Lack of EC-SOD impaired alveolar development and resulted in PH (elevated right ventricular systolic pressures, right ventricular hypertrophy (RVH)), decreased vessel density, and increased small vessel muscularization. Exposure to bleomycin further impaired alveolar development, worsened RVH and vascular remodeling. Lack of EC-SOD and bleomycin treatment decreased lung total and phosphorylated VEGFR2 and eNOS protein expression. CONCLUSION: EC-SOD is critical in preserving normal lung development and loss of EC-SOD results in disrupted alveolar development, PAH and vascular remodeling at baseline, which is further worsened with bleomycin and associated with decreased activation of VEGFR2.


Subject(s)
Bleomycin , Bronchopulmonary Dysplasia/enzymology , Endothelial Cells/enzymology , Hypertension, Pulmonary/enzymology , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/enzymology , Pulmonary Artery/enzymology , Superoxide Dismutase/deficiency , Vascular Remodeling , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/physiopathology , Endothelial Cells/pathology , Genetic Predisposition to Disease , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Phenotype , Phosphorylation , Pulmonary Alveoli/pathology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Signal Transduction , Superoxide Dismutase/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/enzymology , Ventricular Dysfunction, Right/genetics , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right , Ventricular Pressure
15.
Am J Physiol Lung Cell Mol Physiol ; 309(7): L710-24, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26232299

ABSTRACT

The gasotransmitter hydrogen sulfide (H2S) is emerging as a mediator of lung physiology and disease. Recent studies revealed that H2S administration limited perturbations to lung structure in experimental animal models of bronchopulmonary dysplasia (BPD), partially restoring alveolarization, limiting pulmonary hypertension, limiting inflammation, and promoting epithelial repair. No studies have addressed roles for endogenous H2S in lung development. H2S is endogenously generated by cystathionine ß-synthase (Cbs) and cystathionine γ-lyase (Cth). We demonstrate here that the expression of Cbs and Cth in mouse lungs is dynamically regulated during lung alveolarization and that alveolarization is blunted in Cbs(-/-) and Cth(-/-) mouse pups, where a 50% reduction in the total number of alveoli was observed, without any impact on septal thickness. Laser-capture microdissection and immunofluorescence staining indicated that Cbs and Cth were expressed in the airway epithelium and lung vessels. Loss of Cbs and Cth led to a 100-500% increase in the muscularization of small- and medium-sized lung vessels, which was accompanied by increased vessel wall thickness, and an apparent decrease in lung vascular supply. Ablation of Cbs expression using small interfering RNA or pharmacological inhibition of Cth using propargylglycine in lung endothelial cells limited angiogenic capacity, causing a 30-40% decrease in tube length and a 50% decrease in number of tubes formed. In contrast, exogenous administration of H2S with GYY4137 promoted endothelial tube formation. These data confirm a key role for the H2S-generating enzymes Cbs and Cth in pulmonary vascular development and homeostasis and in lung alveolarization.


Subject(s)
Cystathionine beta-Synthase/biosynthesis , Cystathionine gamma-Lyase/biosynthesis , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Hydrogen Sulfide/metabolism , Pulmonary Alveoli , Respiratory Mucosa , Animals , Cystathionine beta-Synthase/genetics , Cystathionine gamma-Lyase/genetics , Mice , Mice, Knockout , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/embryology , Pulmonary Alveoli/enzymology , Respiratory Mucosa/blood supply , Respiratory Mucosa/embryology , Respiratory Mucosa/enzymology
16.
Rev. patol. respir ; 18(2): 82-85, abr.-jun. 2015. ilus
Article in Spanish | IBECS | ID: ibc-141198

ABSTRACT

La presencia de infiltrados pulmonares bilaterales es un hallazgo radiológico frecuente en la práctica clínica habitual. En un varón de edad media no siempre es indicativo de infección y hay que considerar otras entidades diagnósticas (procesos no infecciosos). La sospecha de metástasis pulmonares debe incluirse dentro de estas entidades, aunque se trate de un diagnóstico clínico poco frecuente. La clínica es variable y depende del tumor primario, la edad del paciente, la situación cardiorrespiratoria del mismo y del mecanismo de diseminación tumoral. La linfangitis carcinomatosa es un patrón poco frecuente de metástasis pulmonares y el diagnóstico diferencial se realiza con procesos infecciosos (como neumonías atípicas) e inflamatorios (neumonía intersticial, neumonitis por hipersensibilidad o eosinofilias pulmonares). El paciente que se presenta debutó con clínica respiratoria e infiltrados pulmonares bilaterales y fue diagnosticado de linfangitis carcinomatosa, cuyo origen era un adenocarcinoma gástrico


The presence of bilateral pulmonary infiltrates is a frequent radiological finding in routine clinical practice. In a middle-aged man is not always indicative of infection and you need to consider other diagnostic entities (non-infectious processes). Suspected pulmonary metastases should be included in these entities, although it is a rare clinical diagnosis. The clinical course is variable and depends on the primary tumor, the patient’s age, cardiopulmonary status thereof and the mechanism of tumor dissemination . Carcinomatous lymphangitis is a rare radiographic pattern of pulmonary metastases and the differential diagnosis includes infectious processes (such as atypical pneumonia) and inflammatory diseases (interstitial pneumonia, hypersensitivity pneumonitis or pulmonary eosinophilia). We report the case of a man with respiratory symptoms and bilateral pulmonary infiltrates who was diagnosed of carcinomatous lymphangitis, whose origin was a gastric adenocarcinoma


Subject(s)
Humans , Male , Infiltration-Percolation/methods , Pulmonary Alveoli/abnormalities , Pulmonary Alveoli/pathology , Neoplasms/congenital , Neoplasms/genetics , Lymphangitis/pathology , Therapeutics/instrumentation , Infiltration-Percolation/prevention & control , Pulmonary Alveoli/cytology , Pulmonary Alveoli/enzymology , Neoplasms/complications , Neoplasms/metabolism , Lymphangitis/metabolism , Therapeutics/methods
17.
Biochem Biophys Res Commun ; 459(4): 699-705, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25769947

ABSTRACT

Occurrence of oxidative stress is common in influenza, and renders the host more susceptible to pathogenic effects including cell death. We previously reported that down-regulation of superoxide anion dismutase 1 (SOD1) by influenza A virus (IAV) resulted in a significant increase in the levels of reactive oxygen species (ROS) and viral PB1 polymerase gene product in the early stage of infection. However, the precise molecular mechanism of IAV-mediated ROS generation is not yet fully understood. In this study, we investigated the possible involvement of the key virulence factor PB1-F2 in ROS generation and its contribution to the viral propagation and cell death. The key virulence factor PB1-F2 was found to be responsible, at least in part, for the ROS generation through lowering the SOD1 level in alveolar epithelial A549 cells. PB1-F2 overexpression resulted in SOD1 diminishment and ROS enhancement, while another virulent factor, NS1, did not show significant changes. Inversely, we examined the effects of the absence of PB1-F2 using mutant IAV lacking PB1-F2 expression (mutantΔF2). Infection with mutantΔF2 virus did not significantly lower the SOD1 level, and thus generated moderately low levels of ROS. In addition, the oxidative activity of PB1-F2 was directly reflected by cell viability and death. Infection with the mutant virus reduced the percentage of apoptotic cells more than two-fold compared to the wild-type IAV in A549 cells. Furthermore, expression of exogenous SOD1 gene abrogated a large portion of the PB1-F2-induced apoptosis of cells infected with wild-type IAV, but affected much less of the mutantΔF2 virus-infected cells. These results suggest that the PB1-F2 is directly implicated in virus-induced oxidative stress, thereby contributing to the early stages of IAV replication cycle and ultimately to disease severity.


Subject(s)
Pulmonary Alveoli/metabolism , Viral Proteins/physiology , Animals , Cell Line , Dogs , Humans , Madin Darby Canine Kidney Cells , Oxidation-Reduction , Pulmonary Alveoli/cytology , Pulmonary Alveoli/enzymology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
18.
Immunobiology ; 220(3): 382-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534398

ABSTRACT

BACKGROUND AND OBJECTIVES: Pulmonary alveolar proteinosis (PAP) is characterized by alveolar accumulation of surfactant lipoproteins. Proteasomes are involved in the nonlysosomal protein degradation. We hypothesize that enzymatically active proteasome is increased in the alveolar space of PAP. PATIENTS AND METHODS: 31 PAP patients (29 with primary, 2 with secondary form), 14 disease controls (10 with COPD and 4 with emphysema) and 18 healthy controls were studied. 20S Proteasome was measured by ELISA in bronchoalveolar lavage fluid (BALF) and in serum. Enzyme activity of extracellular proteasome (pkat/mg) was measured through fluorogenic substrate cleavage. RESULTS: Proteasome concentration in BAL was higher in PAP patients than in disease controls or healthy subjects (566±420 vs 53±27 vs 60±42ng/ml, respectively, p<0.0001 for both). Serum proteasome levels were higher in PAP patients than in healthy controls (825±712 vs 405±176ng/ml, p=0.018). PAP patients with active disease had higher serum levels than those who achieved remission (1317±1176 vs 439±422ng/ml, p=0.008). Proteasomal enzyme activity was increased in BAL of PAP patients (p<0.05). CONCLUSIONS: The 20S proteasome is increased and active in BAL of patients with PAP. Extracellular proteasome may contribute to the alveolar degradation of accumulated proteins in PAP.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Proteasome Endopeptidase Complex/blood , Proteasome Endopeptidase Complex/metabolism , Pulmonary Alveolar Proteinosis/pathology , Bronchoalveolar Lavage , Emphysema/metabolism , Female , Humans , Lipoproteins/metabolism , Male , Middle Aged , Proteolysis , Pulmonary Alveoli/enzymology , Pulmonary Disease, Chronic Obstructive/metabolism
19.
Respir Med ; 108(10): 1534-41, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25269709

ABSTRACT

BACKGROUND: In sarcoidosis, an antigen specific immune response is a putative mechanism, resulting in granulomatous inflammation. Since the proteasome is involved in antigen presentation, alterations in the alveolar and parenchymal proteasomal system may be a feature of sarcoidosis. OBJECTIVES: To explore the role of proteasomes and immunoproteasomes in sarcoidosis. METHODS: Total proteasome concentration and activity was assessed in bronchoalveolar lavage (BAL) supernatant obtained from sarcoidosis patients (n = 67) and healthy controls (n = 18) using ELISA and cleavage of specific fluorogenic substrates (±epoxomicin), respectively. Immunohistochemistry of lung tissue sections and immunocytochemistry of BAL macrophages for immunoproteasome was performed in sarcoidosis patients and controls. RESULTS: Proteasome was present in BAL supernatants of all sarcoidosis patients. In sarcoidosis, abundant immunoproteasome staining was seen in pneumocytes type II and granulomas. Total proteasome concentration was greater in active sarcoidosis, stages II (101 ng/ml ± 79; p = 0.009) and III (119 ng/ml ± 66; p = 0.012), than in inactive sarcoidosis or in healthy controls (35 ng/ml ± 34). In the absence of epoxomicin, all fluorogenic substrates were hydrolyzed by BAL supernatant of sarcoidosis patients and controls. CONCLUSIONS: Patients with active sarcoidosis but not healthy controls demonstrate immunoproteasome in the lung tissue and in granulomas. Thus, the putative immune response in sarcoidosis may be mediated or sustained by the proteasomal system.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Granuloma/enzymology , Macrophages, Alveolar/enzymology , Proteasome Endopeptidase Complex/metabolism , Pulmonary Alveoli/enzymology , Sarcoidosis, Pulmonary/enzymology , Adult , Aged , Alveolar Epithelial Cells/enzymology , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
20.
Am J Respir Crit Care Med ; 190(12): 1402-12, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25358054

ABSTRACT

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a deadly lung disease with few therapeutic options. Apoptosis of alveolar epithelial cells, followed by abnormal tissue repair characterized by hyperplastic epithelial cell formation, is a pathogenic process that contributes to the progression of pulmonary fibrosis. However, the signaling pathways responsible for increased proliferation of epithelial cells remain poorly understood. OBJECTIVES: To investigate the role of deoxycytidine kinase (DCK), an important enzyme for the salvage of deoxynucleotides, in the progression of pulmonary fibrosis. METHODS: DCK expression was examined in the lungs of patients with IPF and mice exposed to bleomycin. The regulation of DCK expression by hypoxia was studied in vitro and the importance of DCK in experimental pulmonary fibrosis was examined using a DCK inhibitor and alveolar epithelial cell-specific knockout mice. MEASUREMENTS AND MAIN RESULTS: DCK was elevated in hyperplastic alveolar epithelial cells of patients with IPF and in mice exposed to bleomycin. Increased DCK was localized to cells associated with hypoxia, and hypoxia directly induced DCK in alveolar epithelial cells in vitro. Hypoxia-induced DCK expression was abolished by silencing hypoxia-inducible factor 1α and treatment of bleomycin-exposed mice with a DCK inhibitor attenuated pulmonary fibrosis in association with decreased epithelial cell proliferation. Furthermore, DCK expression, and proliferation of epithelial cells and pulmonary fibrosis was attenuated in mice with conditional deletion of hypoxia-inducible factor 1α in the alveolar epithelium. CONCLUSIONS: Our findings suggest that the induction of DCK after hypoxia plays a role in the progression of pulmonary fibrosis by contributing to alveolar epithelial cell proliferation.


Subject(s)
Deoxycytidine Kinase/physiology , Hypoxia/complications , Idiopathic Pulmonary Fibrosis/etiology , Animals , Cell Line , Cell Proliferation/physiology , Humans , Hypoxia/enzymology , Hypoxia/physiopathology , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/physiopathology , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...