Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
ACS Infect Dis ; 8(7): 1207-1217, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35731709

ABSTRACT

The recruitment of eosinophils into Leishmania lesions is frequently associated with a favorable evolution. A feasible effector for this process is eosinophil cationic protein (ECP, RNase 3), one of the main human eosinophil granule proteins, endowed with a broad spectrum of antimicrobial activity, including parasites. ECP was active on Leishmania promastigotes and axenic amastigotes (LC50's = 3 and 16 µM, respectively) but, in contrast to the irreversible membrane damage caused on bacteria and reproduced by its N-terminal peptides, it only induced a mild and transient plasma membrane destabilization on Leishmania donovani promastigotes. To assess the contribution of RNase activity to the overall leishmanicidal activity of ECP, parasites were challenged in parallel with a single-mutant version, ECP-H15A, devoid of RNase activity, that fully preserves the conformation and liposome permeabilization ability. ECP-H15A showed a similar uptake to ECP on promastigotes, but with higher LC50's (>25 µM) for both parasite stages. ECP-treated promastigotes showed a degraded RNA pattern, absent in ECP-H15A-treated samples. Moreover ECP, but not ECP-H15A, reduced more than 2-fold the parasite burden of infected macrophages. Altogether, our results suggest that ECP enters the Leishmania cytoplasm by an endocytic pathway, ultimately leading to RNA degradation as a key contribution to the leishmanicidal mechanism. Thus, ECP combines both membrane destabilization and enzymatic activities to effect parasite killing. Taken together, our data highlight the microbicidal versatility of ECP as an innate immunity component and support the development of cell-penetrating RNases as putative leishmanicidal agents.


Subject(s)
Anti-Infective Agents , Leishmania donovani , Anti-Infective Agents/pharmacology , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/genetics , Eosinophil Cationic Protein/metabolism , Eosinophil Granule Proteins/pharmacology , Humans , Ribonucleases/metabolism , Ribonucleases/pharmacology
2.
J Med Chem ; 64(15): 11472-11482, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34342438

ABSTRACT

Multidrug resistance against conventional antibiotics poses an important threat to human health. In this context, antimicrobial peptides (AMPs) have been extensively studied for their antibacterial activity and promising results have been shown so far. However, AMPs tend to be rather vulnerable to protease degradation, which offsets their therapeutic appeal. Here, we demonstrate how replacing functional residues in the antimicrobial region of human RNase 3-also named eosinophil cationic protein-by non-natural amino acids increases stability in human serum. These changes were also shown to reduce the hemolytic effect of the peptides in general terms, whereas the antimicrobial activity was reasonably preserved. Digestion profiles enabled us to design new peptides with superior stability and lower toxicity that could become relevant candidates to reach clinical stages.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Eosinophil Cationic Protein/chemistry , Gram-Negative Bacteria/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemical synthesis , Antimicrobial Cationic Peptides/chemistry , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
3.
Cell Mol Life Sci ; 78(6): 2963-2985, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33226440

ABSTRACT

The human RNase3 is a member of the RNaseA superfamily involved in host immunity. RNase3 is expressed by leukocytes and shows broad-spectrum antimicrobial activity. Together with a direct antimicrobial action, RNase3 exhibits immunomodulatory properties. Here, we have analysed the transcriptome of macrophages exposed to the wild-type protein and a catalytic-defective mutant (RNase3-H15A). The analysis of differently expressed genes (DEGs) in treated THP1-derived macrophages highlighted a common pro-inflammatory "core-response" independent of the protein ribonucleolytic activity. Network analysis identified the epidermal growth factor receptor (EGFR) as the main central regulatory protein. Expression of selected DEGs and MAPK phosphorylation were inhibited by an anti-EGFR antibody. Structural analysis suggested that RNase3 activates the EGFR pathway by direct interaction with the receptor. Besides, we identified a subset of DEGs related to the protein ribonucleolytic activity, characteristic of virus infection response. Transcriptome analysis revealed an early pro-inflammatory response, not associated to the protein catalytic activity, followed by a late activation in a ribonucleolytic-dependent manner. Next, we demonstrated that overexpression of macrophage endogenous RNase3 protects the cells against infection by Mycobacterium aurum and the human respiratory syncytial virus. Comparison of cell infection profiles in the presence of Erlotinib, an EGFR inhibitor, revealed that the receptor activation is required for the antibacterial but not for the antiviral protein action. Moreover, the DEGs related and unrelated to the protein catalytic activity are associated to the immune response to bacterial and viral infection, respectively. We conclude that RNase3 modulates the macrophage defence against infection in both catalytic-dependent and independent manners.


Subject(s)
Eosinophil Cationic Protein/metabolism , Amino Acid Sequence , Cell Line , Down-Regulation , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Immunity, Innate , Macrophages/cytology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/virology , Mutagenesis, Site-Directed , Mycobacteriaceae/drug effects , Mycobacteriaceae/physiology , Protein Interaction Maps , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/physiology , Sequence Alignment , Signal Transduction , Transcriptome , Up-Regulation
4.
Int J Mol Sci ; 20(18)2019 Sep 14.
Article in English | MEDLINE | ID: mdl-31540052

ABSTRACT

Candida albicans is a polymorphic fungus responsible for mucosal and skin infections. Candida cells establish themselves into biofilm communities resistant to most currently available antifungal agents. An increase of severe infections ensuing in fungal septic shock in elderly or immunosuppressed patients, along with the emergence of drug-resistant strains, urge the need for the development of alternative antifungal agents. In the search for novel antifungal drugs our laboratory demonstrated that two human ribonucleases from the vertebrate-specific RNaseA superfamily, hRNase3 and hRNase7, display a high anticandidal activity. In a previous work, we proved that the N-terminal region of the RNases was sufficient to reproduce most of the parental protein bactericidal activity. Next, we explored their potency against a fungal pathogen. Here, we have tested the N-terminal derived peptides that correspond to the eight human canonical RNases (RN1-8) against planktonic cells and biofilms of C. albicans. RN3 and RN7 peptides displayed the most potent inhibitory effect with a mechanism of action characterized by cell-wall binding, membrane permeabilization and biofilm eradication activities. Both peptides are able to eradicate planktonic and sessile cells, and to alter their gene expression, reinforcing its role as a lead candidate to develop novel antifungal and antibiofilm therapies.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Peptides/chemistry , Peptides/pharmacology , Ribonucleases/chemistry , Antifungal Agents/chemistry , Biofilms/drug effects , Candida albicans/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Wall/drug effects , Cell Wall/metabolism , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/metabolism , Eosinophil Cationic Protein/pharmacology , Humans , Peptides/metabolism , Ribonucleases/metabolism , Ribonucleases/pharmacology
5.
Sci Rep ; 7(1): 12352, 2017 09 27.
Article in English | MEDLINE | ID: mdl-28955044

ABSTRACT

Cell penetrating peptide derived from human eosinophil cationic protein (CPPecp) is a 10-amino-acid peptide containing a core heparan sulfate (HS)-binding motif of human eosinophil cationic protein (ECP). It binds and penetrates bronchial epithelial cells without cytotoxic effects. Here we investigated airway-protective effects of CPPecp in BEAS-2B cell line and mite-induced airway allergic inflammation in BALB/c mice. In BEAS-2B cell, CPPecp decreases ECP-induced eotaxin mRNA expression. CPPecp also decreases eotaxin secretion and p-STAT6 activation induced by ECP, as well as by IL-4. In vivo studies showed CPPecp decreased mite-induced airway inflammation in terms of eosinophil and neutrophil count in broncho-alveolar lavage fluid, peri-bronchiolar and alveolar pathology scores, cytokine production in lung protein extract including interleukin (IL)-5, IL-13, IL-17A/F, eotaxin; and pause enhancement from methacholine stimulation. CPPecp treated groups also showed lower serum mite-specific IgE level. In this study, we have demonstrated the in vitro and in vivo anti-asthma effects of CPPecp.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Cell-Penetrating Peptides/pharmacology , Eosinophil Cationic Protein/chemistry , Respiratory Mucosa/drug effects , Allergens/immunology , Animals , Anti-Asthmatic Agents/therapeutic use , Antigens, Dermatophagoides/immunology , Asthma/immunology , Asthma/pathology , Bronchi/cytology , Bronchi/drug effects , Bronchi/immunology , Cell Line , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/therapeutic use , Cytokines/immunology , Cytokines/metabolism , Drug Evaluation, Preclinical , Eosinophils/immunology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Treatment Outcome
6.
FEBS J ; 283(22): 4176-4191, 2016 11.
Article in English | MEDLINE | ID: mdl-27696685

ABSTRACT

Acute infection by Gram-negative pathogens can induce an exacerbated immune response that leads to lethal septic shock syndrome. Bacterial lipopolysaccharide (LPS) is a major pathogen-associated molecular pattern molecule that can initiate massive and lethal immune system stimulation. Therefore, the development of new and effective LPS-neutralizing agents is a top priority. The eosinophil cationic protein (ECP) is an antimicrobial protein secreted in response to infection, with a remarkable affinity for LPS. In the present study, we demonstrate that ECP is able to neutralize bacterial LPS and inhibit tumor necrosis factor-α production in human macrophages. We also characterized ECP neutralizing activity using progressively truncated LPS mutants, and conclude that the polysaccharide moiety and lipid A portions are required for LPS-mediated neutralization. In addition, we mapped the structural determinants required for the ECP-LPS interaction by nuclear magnetic resonance. Our results show that ECP is able to neutralize LPS and therefore opens a new route for developing novel therapeutic agents based on the ECP structural scaffolding.


Subject(s)
Endotoxins/metabolism , Eosinophil Cationic Protein/metabolism , Lipopolysaccharides/metabolism , Macrophages/metabolism , Amino Acid Sequence , Cell Line, Tumor , Endotoxins/chemistry , Endotoxins/pharmacology , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/pharmacology , Humans , Kinetics , Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Domains , Protein Structure, Secondary , Sequence Homology, Amino Acid , Spectrometry, Fluorescence , Thermodynamics , Tumor Necrosis Factor-alpha/metabolism
7.
Antimicrob Agents Chemother ; 60(10): 6313-25, 2016 10.
Article in English | MEDLINE | ID: mdl-27527084

ABSTRACT

Eradication of established biofilm communities of pathogenic Gram-negative species is one of the pending challenges for the development of new antimicrobial agents. In particular, Pseudomonas aeruginosa is one of the main dreaded nosocomial species, with a tendency to form organized microbial communities that offer an enhanced resistance to conventional antibiotics. We describe here an engineered antimicrobial peptide (AMP) which combines bactericidal activity with a high bacterial cell agglutination and lipopolysaccharide (LPS) affinity. The RN3(5-17P22-36) peptide is a 30-mer derived from the eosinophil cationic protein (ECP), a host defense RNase secreted by eosinophils upon infection, with a wide spectrum of antipathogen activity. The protein displays high biofilm eradication activity that is not dependent on its RNase catalytic activity, as evaluated by using an active site-defective mutant. On the other hand, the peptide encompasses both the LPS-binding and aggregation-prone regions from the parental protein, which provide the appropriate structural features for the peptide's attachment to the bacterial exopolysaccharide layer and further improved removal of established biofilms. Moreover, the peptide's high cationicity and amphipathicity promote the cell membrane destabilization action. The results are also compared side by side with other reported AMPs effective against either planktonic and/or biofilm forms of Pseudomonas aeruginosa strain PAO1. The ECP and its derived peptide are unique in combining high bactericidal potency and cell agglutination activity, achieving effective biofilm eradication at a low micromolar range. We conclude that the designed RN3(5-17P22-36) peptide is a promising lead candidate against Gram-negative biofilms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Eosinophil Cationic Protein/chemistry , Lipopolysaccharides/metabolism , Pseudomonas aeruginosa/drug effects , Agglutination/drug effects , Animals , Anti-Bacterial Agents/metabolism , Biofilms/drug effects , Erythrocytes/drug effects , Humans , Microbial Sensitivity Tests , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Plankton/microbiology , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/physiology
8.
Part Fibre Toxicol ; 13(1): 24, 2016 05 06.
Article in English | MEDLINE | ID: mdl-27154411

ABSTRACT

Exposure to diesel exhaust (DE) is known to exacerbate allergic inflammation, including virus-induced eosinophil activation in laboratory animals. We have previously shown that in human volunteers with allergic rhinitis a short-term exposure to DE prior to infection with the live attenuated influenza virus (LAIV) increases markers of allergic inflammation in the nasal mucosa. Specifically, levels of eosinophilic cationic protein (ECP) were significantly enhanced in individuals exposed to DE prior to inoculation with LAIV and this effect was maintained for at least seven days. However, this previous study was limited in its scope of nasal immune endpoints and did not explore potential mechanisms mediating the prolonged exacerbation of allergic inflammation caused by exposure to DE prior to inoculation with LAIV. In this follow-up study, the methods were modified to expand experimental endpoints and explore the potential role of NK cells. The data presented here suggest DE prolongs viral-induced eosinophil activation, which was accompanied by decreased markers of NK cell recruitment and activation. Separate in vitro studies showed that exposure to DE particles decreases the ability of NK cells to kill eosinophils. Taken together, these follow-up studies suggest that DE-induced exacerbation of allergic inflammation in the context of viral infections may be mediated by decreased activity of NK cells and their ability to clear eosinophils.


Subject(s)
Air Pollutants/toxicity , Eosinophils/drug effects , Immunity, Mucosal/drug effects , Inhalation Exposure/adverse effects , Killer Cells, Natural/drug effects , Nasal Mucosa/drug effects , Vehicle Emissions/toxicity , Adult , Antibody-Dependent Cell Cytotoxicity/drug effects , Biomarkers/metabolism , Cell Communication , Cell Line , Cells, Cultured , Coculture Techniques , Cohort Studies , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/metabolism , Eosinophils/immunology , Eosinophils/pathology , Female , Follow-Up Studies , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Male , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Rhinitis, Allergic/immunology , Rhinitis, Allergic/metabolism , Rhinitis, Allergic/pathology , Rhinitis, Allergic/physiopathology , Young Adult
9.
Cytometry A ; 89(6): 601-7, 2016 06.
Article in English | MEDLINE | ID: mdl-27061608

ABSTRACT

The analysis of heterogeneous cell samples by mass cytometry (CyTOF) relies on the assumption that metal labeled antibodies accurately bind to their target antigens. We report a previously unappreciated experimental artifact of non-specific antibody binding by eosinophils during intracellular CyTOF analysis of human whole blood samples. We hypothesized that this non-specific binding results from a charge-based interaction between the metal-labeled antibodies and highly cationic proteins found in eosinophillic granules and found that this non-specific staining artifact could be reduced to background levels with a simple blocking protocol using heparin as a competing anionic protein. This protocol eliminates a potential source of erroneous data interpretation in all experiments involving intracellular staining of human whole blood samples, and allows accurate assessment of dynamic changes in intracellular proteins in eosinophils by CyTOF. © 2016 International Society for Advancement of Cytometry.


Subject(s)
Antibodies/chemistry , Eosinophil Cationic Protein/chemistry , Eosinophils/cytology , Flow Cytometry/standards , Heparin/chemistry , Mass Spectrometry/standards , Staining and Labeling/standards , Antigens/chemistry , Antigens/immunology , Artifacts , Binding, Competitive , Child , Cytoplasmic Granules/chemistry , Cytoplasmic Granules/immunology , Eosinophil Cationic Protein/antagonists & inhibitors , Eosinophils/chemistry , Eosinophils/immunology , Flow Cytometry/instrumentation , Humans , Mass Spectrometry/instrumentation , Primary Cell Culture , Protein Binding , Staining and Labeling/methods
10.
Dalton Trans ; 45(2): 579-90, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26609781

ABSTRACT

A novel platinum(ii) organometallic complex, [Pt(pbi)(Me)(DMSO)], bearing the 2-(2'-pyridyl)-benzimidazole (pbiH) ligand, was synthesized and fully characterized. Interestingly, the reaction of this organometallic platinum(ii) complex with two distinct gold(i) phosphane compounds afforded the corresponding heterobimetallic derivatives with the pbi ligand bridging the two metal centers. The antiproliferative properties in vitro of [Pt(pbi)(Me)(DMSO)] and its gold(i) derivatives as well as those of the known coordination platinum(ii) and palladium(ii) complexes with the same ligand, of the general formula [MCl2(pbiH)], were comparatively evaluated against A2780 cancer cells, either sensitive or resistant to cisplatin. A superior biological activity of the organometallic compound clearly emerged compared to the corresponding platinum(ii) complex; the antiproliferative effects are further enhanced upon attaching the gold(i) triphenylphosphine moiety to the organometallic Pt compound. Remarkably, these novel metal species are able to overcome nearly complete resistance to cisplatin. Significant mechanistic insight into the study compounds was gained after investigating their reactions with a few representative biomolecules by electrospray mass spectrometry and X-ray crystallography. The obtained results are comprehensively discussed.


Subject(s)
Coordination Complexes/chemistry , Gold/chemistry , Platinum/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Crystallography, X-Ray , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/metabolism , Humans , Inhibitory Concentration 50 , Molecular Dynamics Simulation , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet
11.
Biomed Res Int ; 2015: 237969, 2015.
Article in English | MEDLINE | ID: mdl-26064887

ABSTRACT

As heparan sulfate proteoglycans (HSPGs) are known as co-receptors to interact with numerous growth factors and then modulate downstream biological activities, overexpression of HS/HSPG on cell surface acts as an increasingly reliable prognostic factor in tumor progression. Cell penetrating peptides (CPPs) are short-chain peptides developed as functionalized vectors for delivery approaches of impermeable agents. On cell surface negatively charged HS provides the initial attachment of basic CPPs by electrostatic interaction, leading to multiple cellular effects. Here a functional peptide (CPPecp) has been identified from critical HS binding region in hRNase3, a unique RNase family member with in vitro antitumor activity. In this study we analyze a set of HS-binding CPPs derived from natural proteins including CPPecp. In addition to cellular binding and internalization, CPPecp demonstrated multiple functions including strong binding activity to tumor cell surface with higher HS expression, significant inhibitory effects on cancer cell migration, and suppression of angiogenesis in vitro and in vivo. Moreover, different from conventional highly basic CPPs, CPPecp facilitated magnetic nanoparticle to selectively target tumor site in vivo. Therefore, CPPecp could engage its capacity to be developed as biomaterials for diagnostic imaging agent, therapeutic supplement, or functionalized vector for drug delivery.


Subject(s)
Cell Movement/drug effects , Cell-Penetrating Peptides/administration & dosage , Heparitin Sulfate/administration & dosage , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell-Penetrating Peptides/chemistry , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/metabolism , Humans , Mice , Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
J Biol Chem ; 290(14): 8863-75, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25713137

ABSTRACT

RNase A is the prototype of an extensive family of divergent proteins whose members share a unique disulfide-bonded tertiary structure, conserved catalytic motifs, and the ability to hydrolyze polymeric RNA. Several members of this family maintain independent roles as ribonucleases and modulators of innate immunity. Here we characterize mouse eosinophil-associated RNase (Ear) 11, a divergent member of the eosinophil ribonuclease cluster, and the only known RNase A ribonuclease expressed specifically in response to Th2 cytokine stimulation. Mouse Ear 11 is differentially expressed in somatic tissues at baseline (brain ≪ liver < lung < spleen); systemic stimulation with IL-33 results in 10-5000-fold increased expression in lung and spleen, respectively. Ear 11 is also expressed in response to protective priming of the respiratory mucosa with Lactobacillus plantarum; transcripts are detected both locally in lung as well as systemically in bone marrow and spleen. Mouse Ear 11 is enzymatically active, although substantially less so than mEar 1 and mEar 2; the relative catalytic efficiency (kcat/Km) of mEar 11 is diminished ∼1000-1500-fold. However, in contrast to RNase 2/EDN and mEar 2, which have been characterized as selective chemoattractants for CD11c(+) dendritic cells, mEar 11 has prominent chemoattractant activity for F4/80(+)CD11c(-) tissue macrophages. Chemoattractant activity is not dependent on full enzymatic activity, and requires no interaction with the pattern recognition receptor, Toll-like receptor 2 (TLR2). Taken together, this work characterizes a divergent RNase A ribonuclease with a unique expression pattern and function, and highlights the versatility of this family in promoting innate immunity.


Subject(s)
Eosinophil Cationic Protein/metabolism , Macrophages/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/genetics , Immunity, Innate , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Phylogeny , Sequence Homology, Amino Acid , Spleen/cytology
13.
PLoS One ; 8(12): e82585, 2013.
Article in English | MEDLINE | ID: mdl-24349317

ABSTRACT

Human eosinophil cationic protein (ECP) and eosinophil derived neurotoxin (EDN) are two ribonuclease A (RNaseA) family members secreted by activated eosinophils. They share conserved catalytic triad and similar three dimensional structures. ECP and EDN are heparin binding proteins with diverse biological functions. We predicted a novel molecular model for ECP binding of heparin hexasaccharide (Hep6), [GlcNS(6S)-IdoA(2S)]3, and residues Gln(40), His(64) and Arg(105) were indicated as major contributions for the interaction. Interestingly, Gln(40) and His(64) on ECP formed a clamp-like structure to stabilize Hep6 in our model, which was not observed in the corresponding residues on EDN. To validate our prediction, mutant ECPs including ECP Q40A, H64A, R105A, and double mutant ECP Q40A/H64A were generated, and their binding affinity for heparins were measured by isothermal titration calorimetry (ITC). Weaker binding of ECP Q40A/H64A of all heparin variants suggested that Gln(40)-His(64) clamp contributed to ECP-heparin interaction significantly. Our in silico and in vitro data together demonstrate that ECP uses not only major heparin binding region but also use other surrounding residues to interact with heparin. Such correlation in sequence, structure, and function is a unique feature of only higher primate ECP, but not EDN.


Subject(s)
Eosinophil Cationic Protein/metabolism , Heparin/metabolism , Amino Acid Sequence , Binding Sites , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/genetics , Eosinophils/enzymology , Gene Expression , Heparin/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Oligosaccharides/chemistry , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Thermodynamics
14.
FEBS J ; 280(22): 5841-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23992292

ABSTRACT

The ribonuclease (RNase) A superfamily lineage includes distant members with antimicrobial properties, suggesting a common ancestral host-defense role. In an effort to identify the minimal requirements for the eosinophil cationic protein (ECP or RNase 3) antimicrobial properties we applied site-directed mutagenesis on its closest family homolog, the eosinophil-derived neurotoxin (EDN or RNase 2). Both eosinophil secretion proteins are involved in human immune defense, and are reported as being among the most rapidly evolving coding sequences in primates. Previous studies in our laboratory defined two regions at the N-terminus involved in the protein antimicrobial action, encompassing residues 8-16 and 34-36. Here, we demonstrate that switching two single residues is enough to provide EDN with ECP antipathogen properties. That is, the EDN double-mutant Q34R/R35W displays enhanced bactericidal activity, particularly towards Gram-negative bacteria, and a significant increase in its affinity towards the bacterial outer membrane lipopolysaccharides. Moreover, we confirmed the direct contribution of residue W35 in lipopolysaccharide binding, membrane interaction and permeabilization processes. Furthermore, additional T13 to I substitution provides EDN with an exposed hydrophobic patch required for protein self-aggregation and triggers bacterial agglutination, thereby increasing the final antimicrobial activity by up to 20-fold. Our results highlight how single selected mutations can reshape the entire protein function. This study provides an example of how structure-guided protein engineering can successfully reproduce an evolution selection process towards the emergence of new physiological roles.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Protein Engineering/methods , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/immunology , Agglutinins/genetics , Agglutinins/immunology , Animals , Antimicrobial Cationic Peptides/chemistry , Endoribonucleases/chemistry , Endoribonucleases/genetics , Endoribonucleases/immunology , Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/genetics , Eosinophil Cationic Protein/immunology , Gram-Negative Bacteria/drug effects , Humans , Models, Molecular , Mutagenesis, Site-Directed , Point Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Ribonuclease, Pancreatic/chemistry
15.
PLoS One ; 8(3): e57318, 2013.
Article in English | MEDLINE | ID: mdl-23469189

ABSTRACT

Cell-penetrating peptides (CPPs) are short peptides which can carry various types of molecules into cells; however, although most CPPs rapidly penetrate cells in vitro, their in vivo tissue-targeting specificities are low. Herein, we describe cell-binding, internalization, and targeting characteristics of a newly identified 10-residue CPP, denoted ECP(32-41), derived from the core heparin-binding motif of human eosinophil cationic protein (ECP). Besides traditional emphasis on positively charged residues, the presence of cysteine and tryptophan residues was demonstrated to be essential for internalization. ECP(32-41) entered Beas-2B and wild-type CHO-K1 cells, but not CHO cells lacking of cell-surface glycosaminoglycans (GAGs), indicating that binding of ECP(32-41) to cell-surface GAGs was required for internalization. When cells were cultured with GAGs or pre-treated with GAG-digesting enzymes, significant decreases in ECP(32-41) internalization were observed, suggesting that cell-surface GAGs, especially heparan sulfate proteoglycans were necessary for ECP(32-41) attachment and penetration. Furthermore, treatment with pharmacological agents identified two forms of energy-dependent endocytosis, lipid-raft endocytosis and macropinocytosis, as the major ECP(32-41) internalization routes. ECP(32-41) was demonstrated to transport various cargoes including fluorescent chemical, fluorescent protein, and peptidomimetic drug into cultured Beas-2B cells in vitro, and targeted broncho-epithelial and intestinal villi tissues in vivo. Hence this CPP has the potential to serve as a novel vehicle for intracellular delivery of biomolecules or medicines, especially for the treatment of pulmonary or gastrointestinal diseases.


Subject(s)
Cell-Penetrating Peptides/metabolism , Eosinophil Cationic Protein/chemistry , Epithelial Cells/metabolism , Heparan Sulfate Proteoglycans/metabolism , Membrane Microdomains/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Biological Transport, Active , CHO Cells , Cell Line , Cell-Penetrating Peptides/chemical synthesis , Cricetinae , Cricetulus , Cysteine/chemistry , Cysteine/metabolism , Epithelial Cells/cytology , Heparan Sulfate Proteoglycans/chemistry , Humans , Kinetics , Membrane Microdomains/chemistry , Molecular Sequence Data , Pinocytosis , Protein Binding , Tryptophan/chemistry , Tryptophan/metabolism
16.
Biomed Res Int ; 2013: 170398, 2013.
Article in English | MEDLINE | ID: mdl-23484086

ABSTRACT

Human ribonucleases A (hRNaseA) superfamily consists of thirteen members with high-structure similarities but exhibits divergent physiological functions other than RNase activity. Evolution of hRNaseA superfamily has gained novel functions which may be preserved in a unique region or domain to account for additional molecular interactions. hRNase3 has multiple functions including ribonucleolytic, heparan sulfate (HS) binding, cellular binding, endocytic, lipid destabilization, cytotoxic, and antimicrobial activities. In this study, three putative multifunctional regions, (34)RWRCK(38) (HBR1), (75)RSRFR(79) (HBR2), and (101)RPGRR(105) (HBR3), of hRNase3 have been identified employing in silico sequence analysis and validated employing in vitro activity assays. A heparin binding peptide containing HBR1 is characterized to act as a key element associated with HS binding, cellular binding, and lipid binding activities. In this study, we provide novel insights to identify functional regions of hRNase3 that may have implications for all hRNaseA superfamily members.


Subject(s)
Eosinophil Cationic Protein/chemistry , Models, Chemical , Cell Line , Eosinophil Cationic Protein/metabolism , Heparin/chemistry , Heparin/metabolism , Humans , Lipids/chemistry , Protein Binding , Protein Structure, Tertiary
17.
ACS Chem Biol ; 8(1): 144-51, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23025322

ABSTRACT

Protein-glycosaminoglycan interactions are essential in many biological processes and human diseases, yet how their recognition occurs is poorly understood. Eosinophil cationic protein (ECP) is a cytotoxic ribonuclease that interacts with glycosaminoglycans at the cell surface; this promotes the destabilization of the cellular membrane and triggers ECP's toxic activity. To understand this membrane destabilization event and the differences in the toxicity of ECP and its homologues, the high resolution solution structure of the complex between full length folded ECP and a heparin-derived trisaccharide (O-iPr-α-D-GlcNS6S-α(1-4)-L-IdoA2S-α(1-4)-D-GlcNS6S) has been solved by NMR methods and molecular dynamics simulations. The bound protein retains the tertiary structure of the free protein. The (2)S(0) conformation of the IdoA ring is preferably recognized by the protein. We have identified the precise location of the heparin binding site, dissected the specific interactions responsible for molecular recognition, and defined the structural requirements for this interaction. The structure reveals the contribution of Arg7, Gln14, and His15 in helix α1, Gln40 in strand ß1, His64 in loop 4, and His128 in strand ß6 in the recognition event and corroborates the previously reported participation of residues Arg34-Asn39. The participation of the catalytic triad (His15, Lys38, His128) in recognizing the heparin mimetic reveals, at atomic resolution, the mechanism of heparin's inhibition of ECP's ribonucleolytic activity. We have integrated all the available data to propose a molecular model for the membrane interaction process. The solved NMR complex provides the structural model necessary to design inhibitors to block ECP's toxicity implicated in eosinophil pathologies.


Subject(s)
Eosinophil Cationic Protein/metabolism , Glycosaminoglycans/metabolism , Molecular Dynamics Simulation , Binding Sites , Cell Membrane/chemistry , Cell Membrane/metabolism , Cytotoxins/chemistry , Cytotoxins/metabolism , Eosinophil Cationic Protein/chemistry , Glycosaminoglycans/chemistry , Humans , Magnetic Resonance Spectroscopy , Models, Biological , Models, Molecular , Protein Folding
18.
PLoS Pathog ; 8(11): e1003005, 2012.
Article in English | MEDLINE | ID: mdl-23133388

ABSTRACT

Antimicrobial proteins and peptides (AMPs) are important effectors of the innate immune system that play a vital role in the prevention of infections. Recent advances have highlighted the similarity between AMPs and amyloid proteins. Using the Eosinophil Cationic Protein as a model, we have rationalized the structure-activity relationships between amyloid aggregation and antimicrobial activity. Our results show how protein aggregation can induce bacteria agglutination and cell death. Using confocal and total internal reflection fluorescence microscopy we have tracked the formation in situ of protein amyloid-like aggregates at the bacteria surface and on membrane models. In both cases, fibrillar aggregates able to bind to amyloid diagnostic dyes were detected. Additionally, a single point mutation (Ile13 to Ala) can suppress the protein amyloid behavior, abolishing the agglutinating activity and impairing the antimicrobial action. The mutant is also defective in triggering both leakage and lipid vesicle aggregation. We conclude that ECP aggregation at the bacterial surface is essential for its cytotoxicity. Hence, we propose here a new prospective biological function for amyloid-like aggregates with potential biological relevance.


Subject(s)
Amyloid/chemistry , Anti-Bacterial Agents/chemistry , Bacteria/chemistry , Eosinophil Cationic Protein/chemistry , Immunity, Innate , Microbial Viability , Amino Acid Substitution , Amyloid/genetics , Amyloid/immunology , Anti-Bacterial Agents/immunology , Bacteria/genetics , Bacteria/immunology , Bacterial Infections , Eosinophil Cationic Protein/genetics , Eosinophil Cationic Protein/immunology , Humans , Mutation, Missense
19.
J Biol Chem ; 287(53): 44289-300, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23135272

ABSTRACT

Conformational flexibility between structural ensembles is an essential component of enzyme function. Although the broad dynamical landscape of proteins is known to promote a number of functional events on multiple time scales, it is yet unknown whether structural and functional enzyme homologues rely on the same concerted residue motions to perform their catalytic function. It is hypothesized that networks of contiguous and flexible residue motions occurring on the biologically relevant millisecond time scale evolved to promote and/or preserve optimal enzyme catalysis. In this study, we use a combination of NMR relaxation dispersion, model-free analysis, and ligand titration experiments to successfully capture and compare the role of conformational flexibility between two structural homologues of the pancreatic ribonuclease family: RNase A and eosinophil cationic protein (or RNase 3). In addition to conserving the same catalytic residues and structural fold, both homologues show similar yet functionally distinct clusters of millisecond dynamics, suggesting that conformational flexibility can be conserved among analogous protein folds displaying low sequence identity. Our work shows that the reduced conformational flexibility of eosinophil cationic protein can be dynamically and functionally reproduced in the RNase A scaffold upon creation of a chimeric hybrid between the two proteins. These results support the hypothesis that conformational flexibility is partly required for catalytic function in homologous enzyme folds, further highlighting the importance of dynamic residue sectors in the structural organization of proteins.


Subject(s)
Eosinophil Cationic Protein/chemistry , Eosinophil Cationic Protein/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/metabolism , Adenosine Monophosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Eosinophil Cationic Protein/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Ribonuclease, Pancreatic/genetics
20.
Biol Chem ; 393(8): 801-15, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22944682

ABSTRACT

Antimicrobial RNases are small cationic proteins belonging to the vertebrate RNase A superfamily and endowed with a wide range of antipathogen activities. Vertebrate RNases, while sharing the active site architecture, are found to display a variety of noncatalytical biological properties, providing an excellent example of multitask proteins. The antibacterial activity of distant related RNases suggested that the family evolved from an ancestral host-defence function. The review provides a structural insight into antimicrobial RNases, taking as a reference the human RNase 3, also named eosinophil cationic protein (ECP). A particular high binding affinity against bacterial wall structures mediates the protein action. In particular, the interaction with the lipopolysaccharides at the Gram-negative outer membrane correlates with the protein antimicrobial and specific cell agglutinating activity. Although a direct mechanical action at the bacteria wall seems to be sufficient to trigger bacterial death, a potential intracellular target cannot be discarded. Indeed, the cationic clusters at the protein surface may serve both to interact with nucleic acids and cell surface heterosaccharides. Sequence determinants for ECP activity were screened by prediction tools, proteolysis and peptide synthesis. Docking results are complementing the structural analysis to delineate the protein anchoring sites for anionic targets of biological significance.


Subject(s)
Anti-Bacterial Agents/chemistry , Eosinophil Cationic Protein/chemistry , Immunologic Factors/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/immunology , Bacterial Physiological Phenomena , Eosinophil Cationic Protein/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate , Immunologic Factors/immunology , Models, Molecular , Molecular Sequence Data , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...