Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Sci Rep ; 10(1): 3471, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32103047

ABSTRACT

To develop genetic resources for the improvement of insects as food, we sequenced transcripts from embryos, one-day hatchlings, three nymphal stages, and male and female adults of the house cricket, Acheta domesticus. A draft transcriptome was assembled from more than 138 million sequences combined from all life stages and sexes. The draft transcriptome assembly contained 45,866 contigs, and more than half were similar to sequences at NCBI (e value < e-3). The highest sequence identity was found in sequences from the termites Cryptotermes secundus and Zootermopsis nevadensis. Sequences with identity to Gregarina niphandrodes suggest that these crickets carry the parasite. Among all life stages, there were 5,042 genes with differential expression between life stages (significant at p < 0.05). An enrichment analysis of gene ontology terms from each life stage or sex highlighted genes that were important to biological processes in cricket development. We further characterized genes that may be important in future studies of genetically modified crickets for improved food production, including those involved in RNA interference, and those encoding prolixicin and hexamerins. The data represent an important first step in our efforts to provide genetically improved crickets for human consumption and livestock feed.


Subject(s)
Gryllidae/genetics , Transcriptome , Animals , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Crop Production , Embryo, Nonmammalian/metabolism , Female , Gene Expression Profiling , Gryllidae/growth & development , Gryllidae/metabolism , Insect Proteins/antagonists & inhibitors , Insect Proteins/genetics , Insect Proteins/metabolism , Life Cycle Stages , Male , Nymph/genetics , Nymph/metabolism , RNA Interference
2.
Am J Respir Cell Mol Biol ; 62(3): 354-363, 2020 03.
Article in English | MEDLINE | ID: mdl-31545652

ABSTRACT

Comparisons of infectivity among the clinically important nontuberculous mycobacteria (NTM) species have not been explored in great depth. Rapid-growing mycobacteria, including Mycobacterium abscessus and M. porcinum, can cause indolent but progressive lung disease. Slow-growing members of the M. avium complex are the most common group of NTM to cause lung disease, and molecular approaches can now distinguish between several distinct species of M. avium complex including M. intracellulare, M. avium, M. marseillense, and M. chimaera. Differential infectivity among these NTM species may, in part, account for differences in clinical outcomes and response to treatment; thus, knowing the relative infectivity of particular isolates could increase prognostication accuracy and enhance personalized treatment. Using human macrophages, we investigated the infectivity and virulence of nine NTM species, as well as multiple isolates of the same species. We also assessed their capacity to evade killing by the antibacterial peptide cathelicidin (LL-37). We discovered that the ability of different NTM species to infect macrophages varied among the species and among isolates of the same species. Our biochemical assays implicate modified phospholipids, which may include a phosphatidylinositol or cardiolipin backbone, as candidate antagonists of LL-37 antibacterial activity. The high variation in infectivity and virulence of NTM strains suggests that more detailed microbiological and biochemical characterizations are necessary to increase our knowledge of NTM pathogenesis.


Subject(s)
Antimicrobial Cationic Peptides/antagonists & inhibitors , Immune Evasion/physiology , Membrane Lipids/physiology , Nontuberculous Mycobacteria/pathogenicity , Phospholipids/physiology , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Cell Membrane/immunology , Chromatography, Thin Layer , Escherichia coli/drug effects , Humans , Macrophages/microbiology , Macrophages, Alveolar/microbiology , Membrane Lipids/isolation & purification , Nontuberculous Mycobacteria/drug effects , Nontuberculous Mycobacteria/physiology , Phospholipids/isolation & purification , Phylogeny , Species Specificity , THP-1 Cells , Virulence , Cathelicidins
3.
J Exp Clin Cancer Res ; 38(1): 422, 2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31640758

ABSTRACT

BACKGROUND: Cancer-initiating cell (CIC), a functionally homogeneous stem-like cell population, is resonsible for driving the tumor maintenance and metastasis, and is a source of chemotherapy and radiation-therapy resistance within tumors. Targeting CICs self-renewal has been proposed as a therapeutic goal and an effective approach to control tumor growth. BMI-1, a critical regulator of self-renewal in the maintenance of CICs, is identified as a potential target for colorectal cancer therapy. METHODS: Colorectal cancer stem-like cell lines HCT116 and HT29 were used for screening more than 500 synthetic compounds by sulforhodamine B (SRB) cell proliferation assay. The candidate compound was studied in vitro by SRB cell proliferation assay, western blotting, cell colony formation assay, quantitative real-time PCR, flow cytometry analysis, and transwell migration assay. Sphere formation assay and limiting dilution analysis (LDA) were performed for measuring the effect of compound on stemness properties. In vivo subcutaneous tumor growth xenograft model and liver metastasis model were performed to test the efficacy of the compound treatment. Student's t test was applied for statistical analysis. RESULTS: We report the development and characterization of a small molecule inhibitor QW24 against BMI-1. QW24 potently down-regulates BMI-1 protein level through autophagy-lysosome degradation pathway without affecting the BMI-1 mRNA level. Moreover, QW24 significantly inhibits the self-renewal of colorectal CICs in stem-like colorectal cancer cell lines, resulting in the abrogation of their proliferation and metastasis. Notably, QW24 significantly suppresses the colorectal tumor growth without obvious toxicity in the subcutaneous xenograft model, as well as decreases the tumor metastasis and increases mice survival in the liver metastasis model. Moreover, QW24 exerts a better efficiency than the previously reported BMI-1 inhibitor PTC-209. CONCLUSIONS: Our preclinical data show that QW24 exerts potent anti-tumor activity by down-regulating BMI-1 and abrogating colorectal CICs self-renewal without obvious toxicity in vivo, suggesting that QW24 could potentially be used as an effective therapeutic agent for clinical colorectal cancer treatment.


Subject(s)
Antimicrobial Cationic Peptides/antagonists & inhibitors , Animals , Cell Line, Tumor , Colorectal Neoplasms , Humans , Male , Mice , Mice, Nude , Xenograft Model Antitumor Assays
4.
Br J Dermatol ; 181(2): 314-323, 2019 08.
Article in English | MEDLINE | ID: mdl-30657173

ABSTRACT

BACKGROUND: Biologics targeting inflammatory mediators can achieve clinical improvements in hidradenitis suppurativa (HS). However, their clinical efficacy shows great interpatient variability in daily practice. OBJECTIVES: To investigate the anti-inflammatory potency of a selection of currently available biologics and prednisolone for the treatment of HS in an ex vivo skin culture system using lesional HS biopsies. METHODS: Lesional skin samples from 10 patients with HS and skin samples from five healthy controls were cultured ex vivo and exposed to prednisolone or biologics targeting tumour necrosis factor (TNF)-α, interleukin (IL)-17A, IL-12/23p40 or CD20 (adalimumab, infliximab, secukinumab, ustekinumab and rituximab, respectively). Real-time quantitative polymerase chain reaction and cytokine bead arrays were used to measure the inhibitory effect of the biologics on cytokines and antimicrobial peptides (AMPs). RESULTS: The relative mRNA expression of all tested cytokines and AMPs was significantly downregulated by all anti-inflammatory agents (P < 0·001). The protein production of the proinflammatory cytokines TNF-α, interferon γ, IL-1ß, IL-6 and IL-17A was significantly inhibited by adalimumab, infliximab, ustekinumab, prednisolone (all P < 0·001) and rituximab (P = 0·0071), but not by secukinumab (P = 0·0663). On both mRNA and protein levels, adalimumab, infliximab and prednisolone reduced the levels of a broader mix of individual cytokines than secukinumab, ustekinumab and rituximab. Moreover, a significant inhibitory effect on mRNA expression levels of inflammatory markers in healthy control skin was observed only for TNF-α inhibitors (P < 0·001) and prednisolone (P = 0·0015). CONCLUSIONS: This ex vivo study suggests that TNF-α inhibitors and prednisolone are the most powerful inhibitors of proinflammatory cytokines and AMPs in HS lesional skin, which concurs with our clinical experience in patients with HS.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Biological Products/pharmacology , Hidradenitis Suppurativa/drug therapy , Prednisolone/pharmacology , Skin/drug effects , Adult , Anti-Inflammatory Agents/therapeutic use , Antigens, CD20/immunology , Antigens, CD20/metabolism , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Biological Products/therapeutic use , Biopsy , Female , Healthy Volunteers , Hidradenitis Suppurativa/immunology , Hidradenitis Suppurativa/pathology , Hidradenitis Suppurativa/surgery , Humans , Interleukin-12 Subunit p40/antagonists & inhibitors , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Interleukin-17/metabolism , Male , Middle Aged , Organ Culture Techniques , Prednisolone/therapeutic use , Skin/immunology , Skin/metabolism , Skin/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
5.
mBio ; 9(5)2018 10 30.
Article in English | MEDLINE | ID: mdl-30377277

ABSTRACT

The keystone oral pathogen Porphyromonas gingivalis is associated with severe periodontitis. Intriguingly, this bacterium is known to secrete large amounts of an enzyme that converts peptidylarginine into citrulline residues. The present study was aimed at identifying possible functions of this citrullinating enzyme, named Porphyromonas peptidylarginine deiminase (PPAD), in the periodontal environment. The results show that PPAD is detectable in the gingiva of patients with periodontitis, and that it literally neutralizes human innate immune defenses at three distinct levels, namely bacterial phagocytosis, capture in neutrophil extracellular traps (NETs), and killing by the lysozyme-derived cationic antimicrobial peptide LP9. As shown by mass spectrometry, exposure of neutrophils to PPAD-proficient bacteria reduces the levels of neutrophil proteins involved in phagocytosis and the bactericidal histone H2. Further, PPAD is shown to citrullinate the histone H3, thereby facilitating the bacterial escape from NETs. Last, PPAD is shown to citrullinate LP9, thereby restricting its antimicrobial activity. The importance of PPAD for immune evasion is corroborated in the infection model Galleria mellonella, which only possesses an innate immune system. Together, the present observations show that PPAD-catalyzed protein citrullination defuses innate immune responses in the oral cavity, and that the citrullinating enzyme of P. gingivalis represents a new type of bacterial immune evasion factor.IMPORTANCE Bacterial pathogens do not only succeed in breaking the barriers that protect humans from infection, but they also manage to evade insults from the human immune system. The importance of the present study resides in the fact that protein citrullination is shown to represent a new bacterial mechanism for immune evasion. In particular, the oral pathogen P. gingivalis employs this mechanism to defuse innate immune responses by secreting a protein-citrullinating enzyme. Of note, this finding impacts not only the global health problem of periodontitis, but it also extends to the prevalent autoimmune disease rheumatoid arthritis, which has been strongly associated with periodontitis, PPAD activity, and loss of tolerance against citrullinated proteins, such as the histone H3.


Subject(s)
Immune Evasion , Immunity, Innate/drug effects , Periodontitis/microbiology , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/immunology , Protein-Arginine Deiminases/metabolism , Virulence Factors/metabolism , Adult , Antimicrobial Cationic Peptides/antagonists & inhibitors , Extracellular Traps/drug effects , Female , Gingiva/chemistry , Gingiva/microbiology , Humans , Male , Periodontitis/pathology , Phagocytosis/drug effects , Porphyromonas gingivalis/growth & development , Protein-Arginine Deiminases/analysis , Virulence Factors/analysis
6.
J Microbiol Biotechnol ; 28(12): 2141-2144, 2018 Dec 28.
Article in English | MEDLINE | ID: mdl-30394046

ABSTRACT

Based on previous studies reporting the anti-prion activity of poly-L-lysine and poly-L-arginine, we investigated cationic poly-L-ornithine (PLO), poly-L-histidine (PLH), anionic poly-L-glutamic acid (PLE) and uncharged poly-L-threonine (PLT) in cultured cells chronically infected by prions to determine their anti-prion efficacy. While PLE and PLT did not alter the level of PrPSc, PLO and PLH exhibited potent PrPSc inhibition in ScN2a cells. These results suggest that the anti-prion activity of poly-basic amino acids is correlated with the cationicity of their functional groups. Comparison of anti-prion activity of PLO and PLH proposes that the anti-prion activity of poly-basic amino acids is associated with their acidic cellular compartments.


Subject(s)
Endopeptidases/drug effects , Histidine/antagonists & inhibitors , Peptides/antagonists & inhibitors , PrPSc Proteins/drug effects , PrPSc Proteins/metabolism , Amino Acids, Basic , Antimicrobial Cationic Peptides/antagonists & inhibitors , Cell Line/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Peptide Fragments/antagonists & inhibitors , Prion Diseases/prevention & control , Prions/drug effects , Prions/pathogenicity
7.
Trends Pharmacol Sci ; 39(8): 685-694, 2018 08.
Article in English | MEDLINE | ID: mdl-30037389

ABSTRACT

Liver-expressed antimicrobial peptide 2 (LEAP-2), the endogenous noncompetitive allosteric antagonist of the growth hormone secretagogue receptor 1a (GHSR1a), was recently identified as a key endocrine factor regulating systemic energy metabolism. This antagonist impairs the ability of ghrelin to activate GHSR1a and diminishes ghrelin-induced Ca2+ release in vitro. The physiological relevance of the molecular LEAP-2-GHSR1a interaction was subsequently demonstrated in vivo. LEAP-2 is therefore a promising therapeutic target in the treatment of obesity and other metabolic diseases. Here, we discuss not only the current understanding of LEAP-2 in metabolic regulation, but also the potential of this peptide in the treatment of obesity and other diseases that involve dysregulation of the ghrelin system.


Subject(s)
Ghrelin/metabolism , Hepcidins/metabolism , Receptors, Ghrelin/metabolism , Animals , Antimicrobial Cationic Peptides/agonists , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/agonists , Blood Proteins/antagonists & inhibitors , Blood Proteins/metabolism , Endocrine System/metabolism , Energy Metabolism , Glucose/metabolism , Humans , Neurosecretory Systems/metabolism , Obesity/drug therapy , Obesity/metabolism , Receptors, Ghrelin/agonists , Receptors, Ghrelin/antagonists & inhibitors
8.
Cell Immunol ; 331: 59-66, 2018 09.
Article in English | MEDLINE | ID: mdl-29861070

ABSTRACT

Neuroinflammation plays a pivotal role in the incidence and progression of Alzheimer's disease (AD). Cathelicidin-related antimicrobial peptide (CRAMP) is critically involved in the innate neuronal responses of chronic neuroinflammation in AD and thus plays a key role in the disease. Here, we show that Aß42 induced microglial production of CRAMP, which was effectively inhibited by milk-fat globule-epidermal growth factor 8 (MFG-E8). Production of CRAMP was associated with activation of ERK1/2, p38 and phospho-P65-NF-kB upregulation. Additionally, the phosphorylation of these signaling proteins was also reversed by MFG-E8. Pre-incubation with signaling inhibitors confirmed that MFG-E8 has a regulatory role on CRAMP through MAPK and NF-kB signaling pathways. MFG-E8 treatment may thus be a potential pharmacotherapy for chronic inflammation in AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Antigens, Surface/pharmacology , Antimicrobial Cationic Peptides/antagonists & inhibitors , Microglia/drug effects , Milk Proteins/pharmacology , Peptide Fragments/toxicity , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Animals, Newborn , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Cells, Cultured , Gene Expression/drug effects , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Mice, Inbred C57BL , Microglia/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Cathelicidins
9.
Crit Care Med ; 46(5): e364-e374, 2018 05.
Article in English | MEDLINE | ID: mdl-29406419

ABSTRACT

OBJECTIVES: Our objectives were to determine first whether albumin prevents heparin-binding protein-induced increased endothelial cell permeability and renal cell inflammation and second, whether a plasma heparin-binding protein-to-albumin ratio predicts risk of acute kidney injury, fluid balance, and plasma cytokine levels in septic shock. DESIGN: In vitro human endothelial and renal cell model and observation cohort of septic shock. SETTINGS: Research laboratory and multicenter clinical trial (Vasopressin and Septic Shock Trial). PATIENTS: Adult septic shock (norepinephrine dose > 5 µg/min for > 6 hr). INTERVENTIONS: In vitro: heparin-binding protein (or thrombin) was added with or without albumin to 1) human endothelial cell monolayers to assess permeability and 2) to human renal tubular epithelial cells to assess inflammation. MEASUREMENTS AND MAIN RESULTS: Transendothelial electrical resistance-a marker of permeability-of human endothelial cells was measured using a voltohmmeter. We measured plasma heparin-binding protein-to-albumin ratio and a panel of cytokines in septic shock patients (n = 330) to define an heparin-binding protein-to-albumin ratio that predicts risk of acute kidney injury. Albumin inhibited heparin-binding protein (and thrombin-induced) increased endothelial cell permeability at a threshold concentration of 20-30 g/L but increased renal tubular cell interleukin-6 release. Patients who developed or had worsened acute kidney injury had significantly higher heparin-binding protein-to-albumin ratio (1.6 vs 0.89; p < 0.001) and heparin-binding protein (38.2 vs 20.8 ng/mL; p < 0.001) than patients without acute kidney injury. The highest heparin-binding protein-to-albumin ratio (> 3.05), heparin-binding protein quartiles (> 69.8), and heparin-binding protein > 30 ng/mL were significantly associated with development or worsening of acute kidney injury (p < 0.001) in unadjusted and adjusted analyses and were robust to sensitivity analyses for death as a competing outcome. Heparin-binding protein and heparin-binding protein-to-albumin ratio were directly associated with positive fluid balance (p < 0.001) and with key inflammatory cytokines. Increasing quartiles of heparin-binding protein-to-albumin ratio and heparin-binding protein (but not albumin) were highly significantly associated with days alive and free of acute kidney injury and renal replacement therapy (p < 0.001), vasopressors (p < 0.001), ventilation (p < 0.001), and with 28-day mortality. CONCLUSIONS: Albumin inhibits heparin-binding protein-induced increased human endothelial cell permeability and heparin-binding protein greater than 30 ng/mL and heparin-binding protein-to-albumin ratio greater than 3.01-but not serum albumin-identified patients at increased risk for acute kidney injury in septic shock.


Subject(s)
Antimicrobial Cationic Peptides/antagonists & inhibitors , Blood Proteins/antagonists & inhibitors , Carrier Proteins/antagonists & inhibitors , Serum Albumin/therapeutic use , Shock, Septic/therapy , Acute Kidney Injury/prevention & control , Antimicrobial Cationic Peptides/blood , Carrier Proteins/blood , Cell Line , Cell Membrane Permeability/drug effects , Cytokines/blood , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Male , Middle Aged , Serum Albumin/analysis , Shock, Septic/blood , Treatment Outcome
10.
Respir Res ; 19(1): 5, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29310642

ABSTRACT

BACKGROUND: Epidemiological studies have shown that urban particulate matter (PM) increases the risk of respiratory infection. However, the underlying mechanisms are poorly understood. PM has been postulated to suppress the activation of airway epithelial innate defence in response to infection. METHODS: The effects of PM on antibacterial defence were studied using an in vitro infection model. The levels of antimicrobial peptides were measured using RT-PCR and ELISA. In addition to performing colony-forming unit counts and flow cytometry, confocal microscopy was performed to directly observe bacterial invasion upon PM exposure. RESULTS: We found that PM PM increased bacterial invasion by impairing the induction of ß-defensin-2 (hBD-2), but not the other antimicrobial peptides (APMs) secreted by airway epithelium. PM further increases bacteria-induced ROS production, which is accompanied by an accelerated cell senescence and a decrease in bacteria-induced hBD-2 production, and the antioxidant NAC treatment attenuates these effects. The PM exposure further upregulated the expression of IL-8 but downregulated the expression of IL-13 upon infection. CONCLUSIONS: PM promotes bacterial invasion of airway epithelial cells by attenuating the induction of hBD-2 via an oxidative burst. These findings associate PM with an increased susceptibility to infection. These findings provide insight into the underlying mechanisms regarding the pathogenesis of particulate matter.


Subject(s)
Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Particulate Matter/adverse effects , Pseudomonas aeruginosa , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cellular Senescence/drug effects , Cellular Senescence/physiology , Dose-Response Relationship, Drug , Humans , Oxidative Stress/drug effects , Oxidative Stress/physiology , Respiratory Mucosa/drug effects , beta-Defensins/antagonists & inhibitors , beta-Defensins/metabolism
11.
J Mol Med (Berl) ; 94(6): 667-79, 2016 06.
Article in English | MEDLINE | ID: mdl-26804311

ABSTRACT

UNLABELLED: Electronic (e)-cigarette use is rapidly rising, with 20 % of Americans ages 25-44 now using these drug delivery devices. E-cigarette users expose their airways, cells of host defense, and colonizing bacteria to e-cigarette vapor (EV). Here, we report that exposure of human epithelial cells at the air-liquid interface to fresh EV (vaped from an e-cigarette device) resulted in dose-dependent cell death. After exposure to EV, cells of host defense-epithelial cells, alveolar macrophages, and neutrophils-had reduced antimicrobial activity against Staphylococcus aureus (SA). Mouse inhalation of EV for 1 h daily for 4 weeks led to alterations in inflammatory markers within the airways and elevation of an acute phase reactant in serum. Upon exposure to e-cigarette vapor extract (EVE), airway colonizer SA had increased biofilm formation, adherence and invasion of epithelial cells, resistance to human antimicrobial peptide LL-37, and up-regulation of virulence genes. EVE-exposed SA were more virulent in a mouse model of pneumonia. These data suggest that e-cigarettes may be toxic to airway cells, suppress host defenses, and promote inflammation over time, while also promoting virulence of colonizing bacteria. KEY MESSAGE: Acute exposure to e-cigarette vapor (EV) is cytotoxic to airway cells in vitro. Acute exposure to EV decreases macrophage and neutrophil antimicrobial function. Inhalation of EV alters immunomodulating cytokines in the airways of mice. Inhalation of EV leads to increased markers of inflammation in BAL and serum. Staphylococcus aureus become more virulent when exposed to EV.


Subject(s)
Electronic Nicotine Delivery Systems , Immunity, Innate/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Nicotiana/toxicity , Pneumonia, Bacterial/immunology , Smoke/adverse effects , Animals , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Cell Death/drug effects , Complex Mixtures/toxicity , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , Macrophages, Alveolar/cytology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Mice , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/metabolism , Pneumonia, Bacterial/microbiology , Nicotiana/chemistry , Cathelicidins
12.
J Biochem ; 159(2): 209-16, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26381537

ABSTRACT

Ethrel is the most effective stimuli in prolonging the latex flow that consequently increases yield per tapping. This effect is largely ascribed to the enhanced lutoid stability, which is associated with the decreased release of initiators of rubber particle (RP) aggregation from lutoid bursting. However, the increase in both the bursting index of lutoids and the duration of latex flow after applying ethrel or ethylene gas in high concentrations suggests that a new mechanism needs to be introduced. In this study, a latex allergen Hev b 7-like protein in C-serum was identified by matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI TOF MS). In vitro analysis showed that the protein acted as a universal antagonist of RP aggregating factors from lutoids and C-serum. Ethrel treatment obviously weakened the effect of C-serum on RP aggregation, which was closely associated with the increase in the level of the Hev b 7-like protein and the decrease in the level of the 37 kDa protein, as revealed by sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE), western blotting analysis and antibody neutralization. Thus, the increase of the Hev b 7-like protein level or the ratio of the Hev b 7-like protein to the 37 kDa protein in C-serum should be primarily ascribed to the ethrel-stimulated prolongation of latex flow duration.


Subject(s)
Antigens, Plant/pharmacology , Hevea/drug effects , Hevea/physiology , Latex/chemistry , Organophosphorus Compounds/pharmacology , Plant Proteins/pharmacology , Antimicrobial Cationic Peptides/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/antagonists & inhibitors , Plant Lectins/antagonists & inhibitors
13.
Immunobiology ; 221(2): 245-52, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26358366

ABSTRACT

Glucocorticoids (GCs) have been extensively used as the mainstream treatment for chronic inflammatory disorders. The persistent use of steroids in the past decades and the association with secondary infections warrants for detailed investigation into their effects on the innate immune system and the therapeutic outcome. In this study, we analyse the effect of GCs on antimicrobial polypeptide (AMP) expression. We hypothesize that GC related side effects, including secondary infections are a result of compromised innate immune responses. Here, we show that treatment with dexamethasone (Dex) inhibits basal mRNA expression of the following AMPs; human cathelicidin, human beta defensin 1, lysozyme and secretory leukocyte peptidase 1 in the THP-1 monocytic cell-line (THP-1 monocytes). Furthermore, pre-treatment with Dex inhibits vitamin D3 induced cathelicidin expression in THP-1 monocytes, primary monocytes and in the human bronchial epithelial cell line BCi NS 1.1. We also demonstrate that treatment with the glucocorticoid receptor (GR) inhibitor RU486 counteracts Dex mediated down-regulation of basal and vitamin D3 induced cathelicidin expression in THP-1 monocytes. Moreover, we confirmed the anti-inflammatory effect of Dex. Pre-treatment with Dex inhibits dsRNA mimic poly IC induction of the inflammatory chemokine IP10 (CXCL10) and cytokine IL1B mRNA expression in THP-1 monocytes. These results suggest that GCs inhibit innate immune responses, in addition to exerting beneficial anti-inflammatory effects.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Cholecalciferol/pharmacology , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Glucocorticoids/pharmacology , Macrophages/drug effects , Antimicrobial Cationic Peptides/agonists , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/immunology , Cell Differentiation , Cell Line , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Cholecalciferol/antagonists & inhibitors , Epithelial Cells/cytology , Epithelial Cells/immunology , Gene Expression Regulation , Humans , Immunity, Innate/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/cytology , Macrophages/immunology , Mifepristone/pharmacology , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Muramidase/antagonists & inhibitors , Muramidase/genetics , Muramidase/immunology , Poly I-C/pharmacology , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Signal Transduction , beta-Defensins/antagonists & inhibitors , beta-Defensins/genetics , beta-Defensins/immunology , Cathelicidins
14.
J Biomol Screen ; 21(4): 363-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26701099

ABSTRACT

Patients with severe lung disease may develop hypercapnia, elevation of the levels of CO2 in the lungs and blood, which is associated with increased risk of death, often from infection. To identify compounds that ameliorate the adverse effects of hypercapnia, we performed a focused screen of 8832 compounds using a CO2-responsive luciferase reporter in Drosophila S2* cells. We found that evoxine, a plant alkaloid, counteracts the CO2-induced transcriptional suppression of antimicrobial peptides in S2* cells. Strikingly, evoxine also inhibits hypercapnic suppression of interleukin-6 and the chemokine CCL2 expression in human THP-1 macrophages. Evoxine's effects are selective, since it does not prevent hypercapnic inhibition of phagocytosis by THP-1 cells or CO2-induced activation of AMPK in rat ATII pulmonary epithelial cells. The results suggest that hypercapnia suppresses innate immune gene expression by definable pathways that are evolutionarily conserved and demonstrate for the first time that specific CO2 effects can be targeted pharmacologically.


Subject(s)
Alkaloids/pharmacology , Carbon Dioxide/antagonists & inhibitors , Epithelial Cells/drug effects , High-Throughput Screening Assays , Macrophages/drug effects , Animals , Antimicrobial Cationic Peptides/agonists , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Carbon Dioxide/toxicity , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Drosophila melanogaster/cytology , Drosophila melanogaster/immunology , Epithelial Cells/cytology , Epithelial Cells/immunology , Gene Expression , Genes, Reporter , Humans , Hypercapnia/prevention & control , Interleukin-6/genetics , Interleukin-6/immunology , Luciferases/genetics , Luciferases/metabolism , Macrophages/cytology , Macrophages/immunology
15.
Cell Host Microbe ; 18(4): 390-1, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468741

ABSTRACT

Virulent group A streptococci have become a serious threat, with the emergence of the hypervirulent lineage M1T1. In this issue of Cell Host & Microbe, LaRock et al. (2015), uncover a role for the streptococcal M1 protein in neutralizing a key human antimicrobial peptide, cathelicidin.


Subject(s)
Antigens, Bacterial/metabolism , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Immune Evasion , Streptococcus pyogenes/immunology , Streptococcus pyogenes/metabolism , Animals , Humans
16.
Cell Host Microbe ; 18(4): 471-7, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468750

ABSTRACT

The antimicrobial peptide LL-37 is generated upon proteolytic cleavage of cathelicidin and limits invading pathogens by directly targeting microbial membranes as well as stimulating innate immune cell function. However, some microbes evade LL-37-mediated defense. Notably, group A Streptococcus (GAS) strains belonging to the hypervirulent M1T1 serogroup are more resistant to human LL-37 than other GAS serogroups. We show that the GAS surface-associated M1 protein sequesters and neutralizes LL-37 antimicrobial activity through its N-terminal domain. M1 protein also binds the cathelicidin precursor hCAP-18, preventing its proteolytic maturation into antimicrobial forms. Exogenous M1 protein rescues M1-deficient GAS from killing by neutrophils and within neutrophil extracellular traps and neutralizes LL-37 chemotactic properties. M1 also binds murine cathelicidin, and its virulence contribution in a murine model of necrotizing skin infection is largely driven by its ability to neutralize this host defense peptide. Thus, cathelicidin resistance is essential for the pathogenesis of hyperinvasive M1T1 GAS.


Subject(s)
Antigens, Bacterial/metabolism , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Immune Evasion , Streptococcus pyogenes/immunology , Streptococcus pyogenes/metabolism , Animals , Disease Models, Animal , Humans , Mice , Protein Binding , Skin Diseases, Bacterial/microbiology , Skin Diseases, Bacterial/pathology , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Cathelicidins
17.
Biol Chem ; 396(12): 1369-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26351912

ABSTRACT

Ten secreted aspartic proteases (Saps) of Candida albicans cleave numerous peptides and proteins in the host organism and deregulate its homeostasis. Human kininogens contain two internal antimicrobial peptide sequences, designated NAT26 and HKH20. In our current study, we characterized a Sap-catalyzed cleavage of kininogen-derived antimicrobial peptides that results in the loss of the anticandidal activity of these peptides. The NAT26 peptide was effectively inactivated by all Saps, except Sap10, whereas HKH20 was completely degraded only by Sap9. Proteolytic deactivation of the antifungal potential of human kininogens can help the pathogens to modulate or evade the innate immunity of the host.


Subject(s)
Antimicrobial Cationic Peptides/antagonists & inhibitors , Aspartic Acid Endopeptidases/pharmacology , Candida albicans/enzymology , Host-Pathogen Interactions , Kininogens/metabolism , Amino Acid Sequence , Aspartic Acid Endopeptidases/metabolism , Chromatography, Liquid , Humans , Kininogens/antagonists & inhibitors , Kininogens/chemistry , Molecular Sequence Data
18.
Oncotarget ; 6(5): 2939-50, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25596747

ABSTRACT

Here we found that levels of cathelicidin, an antimicrobial peptide, were increased in colon cancer tissues compared to noncancerous tissues. Importantly, cathelicidin was mainly expressed in immune cells. Contact with tumor cells caused macrophages to secrete cathelicidin. Neutralization of cathelicidin, in vivo, significantly reduced the engraftment of macrophages into colon tumors, as well as proliferation of tumor cells, resulting in an inhibition of tumor growth. Furthermore, treatment with cathelicidin neutralizing antibody de-activated the Wnt/ß-catenin signaling pathway in tumor cells both in vivo and in vitro. Cathelicidin activated Wnt/ß-catenin signaling by inducing phosphorylation of PTEN, leading to activation of PI3K/Akt signaling and subsequent phosphorylation of GSK3ß, resulting in stabilization and nuclear translocation of ß-catenin. These data indicate that cathelicidin, expressed by immune cells in the tumor microenvironment, promotes colon cancer growth through activation of the PTEN/PI3K/Akt and Wnt/ß-catenin signaling pathways.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Cell Transformation, Neoplastic/metabolism , Colitis/complications , Colonic Neoplasms/etiology , Macrophages/metabolism , Paracrine Communication , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/pharmacology , Case-Control Studies , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Coculture Techniques , Colitis/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HCT116 Cells , Humans , Macrophages/drug effects , Mice, Inbred C57BL , PTEN Phosphohydrolase/metabolism , Paracrine Communication/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Time Factors , Transfection , Tumor Burden , Tumor Microenvironment , U937 Cells , Wnt Signaling Pathway/drug effects , Cathelicidins
19.
J Periodontal Res ; 50(1): 80-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24697598

ABSTRACT

BACKGROUND AND OBJECTIVE: High levels of the antimicrobial peptide, LL-37, are detected in gingival crevicular fluid from patients with chronic periodontitis. LL-37 not only shows antimicrobial activity but also affects host-cell viability. The objective of the present study was to identify endogenous mechanisms that antagonize the detrimental effects of LL-37 on osteoblast viability, focusing on the human peptide p33 expressed on the surface of various cell types. MATERIAL AND METHODS: Human osteoblast-like MG63 cells and human hFOB1.19 osteoblasts were treated with or without LL-37 in the presence or absence of p33. Recombinant human p33 was expressed in an Escherichia coli expression system. Lactate dehydrogenase (LDH) was assessed using an enzymatic spectrophotometric assay. DNA synthesis was determined by measuring [(3) H]-thymidine incorporation. Cell number was assessed by counting cells in a Bürker chamber. Intracellular Ca(2+) was monitored by recording Fluo 4-AM fluorescence using a laser scanning confocal microscope. Cellular expression of p33 was determined by western blotting. RESULTS: LL-37 caused a concentration-dependent release of LDH from human osteoblasts, showing a half-maximal response value (EC50 ) of 4 µm and a rapid and sustained rise in the intracellular Ca(2+) concentration of osteoblasts, suggesting that LL-37 forms pores in the cell membrane. p33 (10 µm) inhibited the LL-37-induced LDH release and LL-37-evoked rise in intracellular Ca(2+) concentration, suggesting that p33 prevents LL-37-induced permeabilization of the cell membrane. Moreover, p33 blocked LL-37-induced attenuation of osteoblast numbers. Also, mucin antagonized, at concentrations representative for nonstimulated whole saliva, LL-37-evoked LDH release, whilst cationic endogenous polyamines had no impact on LL-37-induced LDH release from osteoblasts. CONCLUSIONS: The endogenous peptide p33 prevents LL-37-induced reduction of human osteoblast viability. Importantly, this mechanism may protect the osteoblasts from LL-37-induced cell damage in patients suffering from chronic periodontitis associated with high levels of LL-37 locally.


Subject(s)
Antimicrobial Cationic Peptides/antagonists & inhibitors , Complement C1q/pharmacology , Membrane Glycoproteins/pharmacology , Osteoblasts/drug effects , Antimicrobial Cationic Peptides/pharmacology , Calcium/analysis , Carrier Proteins/pharmacology , Cell Count , Cell Line , Cell Membrane/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , DNA/biosynthesis , DNA/drug effects , Humans , L-Lactate Dehydrogenase/analysis , L-Lactate Dehydrogenase/antagonists & inhibitors , Mitochondrial Proteins/pharmacology , Mucins/pharmacology , Receptors, Complement , Cathelicidins
20.
J Biol Chem ; 289(40): 27614-24, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25092290

ABSTRACT

LL-37 is a peptide secreted by human epithelial cells that can lyse bacteria, suppress signaling by Toll-like receptor 4 (TLR4), and enhance signaling to double-stranded RNA (dsRNA) by TLR3. How LL-37 interacts with dsRNA to affect signal transduction by TLR3 is not completely understood. We determined that LL-37 binds dsRNA and traffics to endosomes and releases the dsRNA in a pH-dependent manner. Using dynamic light scattering spectroscopy and cell-based FRET experiments, LL-37 was found to form higher order complexes independent of dsRNA binding. Upon acidification LL-37 will dissociate from a larger complex. In cells, LL-37 has a half-live of ∼ 1 h. LL-37 half-life was increased by inhibiting endosome acidification or inhibiting cathepsins, which include proteases whose activity are activated by endosome acidification. Residues in LL-37 that contact poly(I:C) and facilitate oligomerization in vitro were mapped. Peptide LL-29, which contains the oligomerization region of LL-37, inhibited LL-37 enhancement of TLR3 signal transduction. LL-29 prevented LL-37 · poly(I:C) co-localization to endosomes containing TLR3. These results shed light on the requirements for LL-37 enhancement of TLR3 signaling.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Peptides/chemistry , Signal Transduction , Toll-Like Receptor 3/metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Binding Sites , Endosomes/metabolism , Humans , Hydrogen-Ion Concentration , Molecular Sequence Data , Peptides/metabolism , RNA, Double-Stranded/metabolism , Toll-Like Receptor 3/chemistry , Toll-Like Receptor 3/genetics , Up-Regulation , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL
...