Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Biochemistry ; 52(9): 1547-58, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23406372

ABSTRACT

Cathelicidins form a family of small host defense peptides distinct from another class of cationic antimicrobial peptides, the defensins. They are expressed as large precursor molecules with a highly conserved pro-domain known as the cathelin-like domain (CLD). CLDs have high degrees of sequence homology to cathelin, a protein isolated from pig leukocytes and belonging to the cystatin family of cysteine protease inhibitors. In this report, we describe for the first time the X-ray crystal structure of the human CLD (hCLD) of the sole human cathelicidin, LL-37. The structure of the hCLD, determined at 1.93 Å resolution, shows the cystatin-like fold and is highly similar to the structure of the CLD of the pig cathelicidin, protegrin-3. We assayed the in vitro antibacterial activities of the hCLD, LL-37, and the precursor form, pro-cathelicidin (also known as hCAP18), and we found that the unprocessed protein inhibited the growth of Gram-negative bacteria with efficiencies comparable to that of the mature peptide, LL-37. In addition, the antibacterial activity of LL-37 was not inhibited by the hCLD intermolecularly, because exogenously added hCLD had no effect on the bactericidal activity of the mature peptide. The hCLD itself lacked antimicrobial function and did not inhibit the cysteine protease, cathepsin L. Our results contrast with previous reports of hCLD activity. A comparative structural analysis between the hCLD and the cysteine protease inhibitor stefin A showed why the hCLD is unable to function as an inhibitor of cysteine proteases. In this respect, the cystatin scaffold represents an ancestral structural platform from which proteins evolved divergently, with some losing inhibitory functions.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Animals , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Cathepsin L/antagonists & inhibitors , Crystallography, X-Ray , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Humans , Models, Molecular , Protein Folding , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Swine , Cathelicidins
2.
J Biol Chem ; 287(26): 21615-27, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22573326

ABSTRACT

Human α-defensins are cationic peptides that self-associate into dimers and higher-order oligomers. They bind protein toxins, such as anthrax lethal factor (LF), and kill bacteria, including Escherichia coli and Staphylococcus aureus, among other functions. There are six members of the human α-defensin family: four human neutrophil peptides, including HNP1, and two enteric human defensins, including HD5. We subjected HD5 to comprehensive alanine scanning mutagenesis. We then assayed LF binding by surface plasmon resonance, LF activity by enzyme kinetic inhibition, and antibacterial activity by the virtual colony count assay. Most mutations could be tolerated, resulting in activity comparable with that of wild type HD5. However, the L29A mutation decimated LF binding and bactericidal activity against Escherichia coli and Staphylococcus aureus. A series of unnatural aliphatic and aromatic substitutions at position 29, including aminobutyric acid (Abu) and norleucine (Nle) correlated hydrophobicity with HD5 function. The crystal structure of L29Abu-HD5 depicted decreased hydrophobic contacts at the dimer interface, whereas the Nle-29-HD5 crystal structure depicted a novel mode of dimerization with parallel ß strands. The effect of mutating Leu(29) is similar to that of a C-terminal hydrophobic residue of HNP1, Trp(26). In addition, in order to further clarify the role of dimerization in HD5 function, an obligate monomer was generated by N-methylation of the Glu(21) residue, decreasing LF binding and antibacterial activity against S. aureus. These results further characterize the dimer interface of the α-defensins, revealing a crucial role of hydrophobicity-mediated dimerization.


Subject(s)
alpha-Defensins/physiology , Alanine/chemistry , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Crystallography, X-Ray/methods , Dimerization , Escherichia coli/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Kinetics , Leucine/chemistry , Molecular Conformation , Mutagenesis , Mutation , Peptides/chemistry , Protein Conformation , Staphylococcus aureus/metabolism , Surface Plasmon Resonance , alpha-Defensins/chemistry
3.
J Biol Chem ; 287(23): 18900-12, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22496447

ABSTRACT

The human α-defensins (HNP) are synthesized in vivo as inactive prodefensins, and contain a conserved glycine, Gly(17), which is part of a ß-bulge structure. It had previously been shown that the glycine main chain torsion angles are in a D-configuration, and that d-amino acids but not L-alanine could be substituted at that position to yield correctly folded peptides without the help of a prodomain. In this study, the glycine to L-alanine mutant defensin was synthesized in the form of a prodefensin using native chemical ligation. The ligation product folded correctly and yielded an active peptide upon CNBr cleavage. The L-Ala(17)-HNP1 crystal structure depicted a ß-bulge identical to wild-type HNP1. However, dimerization was perturbed, causing one monomer to tilt with respect to the other in a dimerization model. Inhibitory activity against the anthrax lethal factor showed a 2-fold reduction relative to wild-type HNP1 as measured by the inhibitory concentration IC(50). Self-association was slightly reduced, as detected by surface plasmon resonance measurements. According to the results of the virtual colony count assay, the antibacterial activity against Escherichia coli, Staphylococcus aureus, and Bacillus cereus exhibited a less than 2-fold reduction in virtual lethal dose values. Prodefensins with two other L-amino acid substitutions, Arg and Phe, at the same position did not fold, indicating that only small side chains are tolerable. These results further elucidate the factors governing the region of the ß-bulge structure that includes Gly(17), illuminating why glycine is conserved in all mammalian α-defensins.


Subject(s)
Anti-Infective Agents/chemistry , Glycine/chemistry , Protein Folding , Protein Multimerization , alpha-Defensins/chemistry , Amino Acid Substitution , Anti-Infective Agents/pharmacology , Bacteria/growth & development , Crystallography, X-Ray , Glycine/genetics , Humans , Mutation, Missense , Structure-Activity Relationship , alpha-Defensins/genetics , alpha-Defensins/pharmacology
4.
J Biol Chem ; 287(12): 8944-53, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22270360

ABSTRACT

Human myeloid α-defensins called HNPs play multiple roles in innate host defense. The Trp-26 residue of HNP1 was previously shown to contribute importantly to its ability to kill S. aureus, inhibit anthrax lethal factor (LF), bind gp120 of HIV-1, dimerize, and undergo further self-association. To gain additional insights into the functional significance of dimerization, we compared wild type HNP1 to dimerization-impaired, N-methylated HNP1 monomers and to disulfide-tethered obligate HNP1 dimers. The structural effects of these modifications were confirmed by x-ray crystallographic analyses. Like the previously studied W26A mutation, N-methylation of Ile-20 dramatically reduced the ability of HNP1 to kill Staphylococcus aureus, inhibit LF, and bind gp120. Importantly, this modification had minimal effect on the ability of HNP1 to kill Escherichia coli. The W26A and MeIle-20 mutations impaired defensin activity synergistically. N-terminal covalent tethering rescued the ability of W26A-HNP1 to inhibit LF but failed to restore its defective killing of S. aureus. Surface plasmon resonance studies revealed that Trp-26 mediated the association of monomers and canonical dimers of HNP1 to immobilized HNP1, LF, and gp120, and also indicated a possible mode of tetramerization of HNP1 mediated by Ile-20 and Leu-25. This study demonstrates that dimerization contributes to some but not all of the many and varied activities of HNP1.


Subject(s)
alpha-Defensins/chemistry , alpha-Defensins/immunology , Crystallography, X-Ray , Dimerization , Escherichia coli/physiology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Humans , Immunity, Innate , Molecular Conformation , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , alpha-Defensins/genetics
5.
AAPS J ; 25(6): 103, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37936002

ABSTRACT

The in-person workshop "Drug Dissolution in Oral Drug Absorption" was held on May 23-24, 2023, in Baltimore, MD, USA. The workshop was organized into lectures and breakout sessions. Three common topics that were re-visited by various lecturers were amorphous solid dispersions (ASDs), dissolution/permeation interplay, and in vitro methods to predict in vivo biopharmaceutics performance and risk. Topics that repeatedly surfaced across breakout sessions were the following: (1) meaning and assessment of "dissolved drug," particularly of poorly water soluble drug in colloidal environments (e.g., fed conditions, ASDs); (2) potential limitations of a test that employs sink conditions for a poorly water soluble drug; (3) non-compendial methods (e.g., two-stage or multi-stage method, dissolution/permeation methods); (4) non-compendial conditions (e.g., apex vessels, non-sink conditions); and (5) potential benefit of having both a quality control method for batch release and a biopredictive/biorelevant method for biowaiver or bridging scenarios. An identified obstacle to non-compendial methods is the uncertainty of global regulatory acceptance of such methods.


Subject(s)
Biopharmaceutics , Intestinal Absorption , Humans , Drug Liberation , Solubility , Water
6.
F1000Res ; 11: 933, 2022.
Article in English | MEDLINE | ID: mdl-37360940

ABSTRACT

Background: HNP1, LL-37, and HBD1 are antimicrobial against Escherichia coli ATCC 25922 at the standard inoculum but less active at higher inocula.   Methods: The virtual colony count (VCC) microbiological assay was adapted for high inocula and the addition of yeast tRNA and bovine pancreatic ribonuclease A (RNase).  96-well plates were read for 12 hours in a Tecan Infinite M1000 plate reader and photographed under 10x magnification.    Results: Adding tRNA 1:1 wt/wt to HNP1 at the standard inoculum almost completely abrogated activity.  Adding RNase 1:1 to HNP1 at the standard inoculum of 5x10 5 CFU/mL did not enhance activity.  Increasing the inoculum to 6.25x10 7 CFU/mL almost abrogated HNP1 activity.  However, adding RNase 25:1 to HNP1 enhanced activity at the highest tested concentration of HNP1.  Adding both tRNA and RNase resulted in enhanced activity, indicating that the enhancement effect of RNase overwhelms the inhibiting effect of tRNA when both are present.  HBD1 activity at the standard inoculum was almost completely abrogated by the addition of tRNA, but LL-37 activity was only slightly inhibited by tRNA.  At the high inoculum, LL-37 activity was enhanced by RNase.  HBD1 activity was not enhanced by RNase.  RNase was not antimicrobial in the absence of antimicrobial peptides.  Cell clumps were observed at the high inoculum in the presence of all three antimicrobial peptides and at the standard inoculum in the presence of HNP1+tRNA and HBD1+tRNA.    Conclusions: Antimicrobial peptide-ribonuclease combinations have the potential to be active against high cell concentrations, conditions where the antimicrobial agent alone is relatively ineffective.


Subject(s)
Anti-Infective Agents , Ribonuclease, Pancreatic , Cattle , Animals , Ribonuclease, Pancreatic/pharmacology , Antimicrobial Peptides , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cathelicidins/pharmacology , Escherichia coli
7.
J Biol Chem ; 284(42): 29180-92, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19640840

ABSTRACT

Despite the small size and conserved tertiary structure of defensins, little is known at a molecular level about the basis of their functional versatility. For insight into the mechanism(s) of defensin function, we prepared enantiomeric pairs of four human defensins, HNP1, HNP4, HD5, and HBD2, and studied their killing of bacteria, inhibition of anthrax lethal factor, and binding to HIV-1 gp120. Unstructured HNP1, HD5, and HBD3 and several other human alpha- and beta-defensins were also examined. Crystallographic analysis showed a plane of symmetry that related (L)HNP1 and (D)HNP1 to each other. Either d-enantiomerization or linearization significantly impaired the ability of HNP1 and HD5 to kill Staphylococcus aureus but not Escherichia coli. In contrast, (L)HNP4 and (D)HNP4 were equally bactericidal against both bacteria. d-Enantiomers were generally weaker inhibitors or binders of lethal factor and gp120 than their respective native, all-l forms, although activity differences were modest, particularly for HNP4. A strong correlation existed among these different functions. Our data indicate: (a) that HNP1 and HD5 kill E. coli by a process that is mechanistically distinct from their actions that kill S. aureus and (b) that chiral molecular recognition is not a stringent prerequisite for other functions of these defensins, including their ability to inhibit lethal factor and bind gp120 of HIV-1.


Subject(s)
alpha-Defensins/chemistry , Alanine/chemistry , Aminobutyrates/chemistry , Animals , Antigens, Bacterial/chemistry , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/chemistry , Crystallography, X-Ray/methods , Cysteine/chemistry , Escherichia coli/metabolism , Humans , Kinetics , Mice , Microbial Sensitivity Tests , Staphylococcus aureus/metabolism , Stereoisomerism , Surface Plasmon Resonance
8.
J Pharm Biomed Anal ; 49(2): 240-6, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19070984

ABSTRACT

Cellulose acetate phthalate (CAP, cellulose acetate 1,2-benzenedicarboxylate) is a common polymeric oral tablet coating. CAP is also a vaginal microbicide candidate that potently inhibits HIV-1 proliferation. This paper describes the development of a precise, stability-indicating gel permeation chromatography (GPC) assay for CAP. During accelerated stability studies monitored by separate reversed-phase high performance liquid chromatography (RP-HPLC) and GPC analyses, an apparent loss of mass balance was observed. This deficit was corrected by recalculating the response factor (RF) for each degraded sample, proportional to the fraction of phthalate remaining bound to the polymeric CAP. The correction factor enabled CAP and the degradation product phthalic acid (PA) to be quantitated by a single GPC analysis. The chromatographic approach taken here could potentially apply to any polymer containing degradable chromophores.


Subject(s)
Cellulose/analogs & derivatives , Chromatography, Gel/methods , Anti-HIV Agents/pharmacology , Cellulose/analysis , Cellulose/chemistry , Cellulose/pharmacology , Chromatography, Gel/economics , Chromatography, High Pressure Liquid/methods , Drug Stability , Female , HIV-1/drug effects , Humans , Hydrolysis , Molecular Structure , Reproducibility of Results , Solutions/chemistry , Solvents/chemistry
9.
J Mol Biol ; 368(2): 537-49, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17355880

ABSTRACT

Human neutrophil alpha-defensins (HNPs) are synthesized in vivo as inactive precursor proteins, i.e. preproHNPs. A series of sequential proteolytic events excise the N-terminal inhibitory pro peptide, leading to defensin maturation and storage in azurophilic granules. The anionic pro peptide, required for correct sub-cellular trafficking and sorting of proHNPs, inhibits the antimicrobial activity of cationic defensins, either inter or intra-molecularly, presumably through charge neutralization. To better understand the role of the pro peptide in the folding and functioning of alpha-defensins and/or pro alpha-defensins, we chemically attached the proHNP1 pro peptide or (wt)pro peptide and the following artificial pro segments to the N terminus of HNP1: polyethylene glycol (PEG), Arg(10) (polyR), Ser(10) (polyS), and (cr)pro peptide, a charge-reversing mutant of the pro peptide where Arg/Lys residues were changed to Asp, and Asp/Glu residues to Lys. Comparative in vitro folding suggested that while all artificial pro segments chaperoned defensin folding, with PEG being the most efficient, the pro peptide catalyzed the folding of proHNPs likely through two independent mechanisms: solubilization of and interaction with the C-terminal defensin domain. Further, the N-terminal artificial pro segments dramatically altered the bactericidal activity of HNP1 against both Escherichia coli and Staphylococcus aureus. Surprisingly, (cr)pro peptide and (wt)pro peptide showed similar properties with respect to intra-molecular and inter-molecular catalysis of defensin folding as well as alpha-defensin binding, although their binding modes appeared different. Our findings identify a dual chaperone activity of the pro peptide and may shed light on the molecular mechanisms by which pro alpha-defensins fold in vivo.


Subject(s)
Neutrophils/metabolism , Protein Folding , Protein Precursors/metabolism , alpha-Defensins/chemistry , alpha-Defensins/metabolism , Anti-Bacterial Agents/pharmacology , Catalysis/drug effects , Chromatography, High Pressure Liquid , Escherichia coli/drug effects , Humans , Membranes, Artificial , Microbial Viability/drug effects , Molecular Chaperones/metabolism , Mutant Proteins/metabolism , Neutrophils/drug effects , Oxidation-Reduction/drug effects , Peptides/metabolism , Peptides/pharmacology , Protein Binding/drug effects , Serine/metabolism , Solubility/drug effects , Spectrometry, Fluorescence , Staphylococcus aureus/drug effects , Structure-Activity Relationship , Surface Plasmon Resonance , Time Factors
10.
FEBS Lett ; 579(1): 162-6, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15620707

ABSTRACT

Human neutrophil alpha-defensin 4 (HNP4) is more effective than HNP1-3 in protecting human peripheral blood mononuclear cells from infection by both X4 and R5 HIV-1 strains. HNP4 binds to both CD4 and gp120 approximately two orders of magnitude weaker than does HNP1, and is less effectively sequestered by glycosylated serum proteins than HNP1. These results suggest that the HIV-1 inhibition by HNP4 stems at least partially from a unique and lectin-independent property of HNP4 with CD4 and/or gp120. Our finding identifies an anti-HIV-1 property of HNP4 and may have implications in the development of new antiviral agents for AIDS therapy.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Leukocytes, Mononuclear/virology , alpha-Defensins/pharmacology , Anti-HIV Agents/therapeutic use , CD4 Antigens/metabolism , HIV Envelope Protein gp120/metabolism , HIV Infections/drug therapy , Humans , Ligands , alpha-Defensins/therapeutic use
11.
Peptides ; 26(12): 2377-83, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16009464

ABSTRACT

Human neutrophil alpha-defensin 2 (HNP2) was N-terminally acetylated and/or C-terminally amidated, resulting in three terminally modified analogs, Ac-HNP2, HNP2-NH2 and Ac-HNP2-NH2. We examined their bactericidal activity against E. coli and S. aureus and their ability to induce leakage from large unilamellar vesicles. Loss of the N-terminal positive charge was functionally deleterious, whereas removal of the C-terminal negative charge enhanced microbial killing and membrane permeabilization. Our findings validate the importance of electrostatic forces in defensin-microbe interactions and point to the bacterial cytoplasmic membrane as a target of HNP2 activity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/growth & development , Staphylococcus aureus/growth & development , alpha-Defensins/pharmacology , Acetylation , Anti-Bacterial Agents/chemistry , Cell Membrane Permeability/drug effects , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Protein Conformation , Protein Structure, Secondary , alpha-Defensins/chemistry
12.
F1000Res ; 4: 1, 2015.
Article in English | MEDLINE | ID: mdl-29333228

ABSTRACT

Dark blue rings and circles emerged when the non-specific polysaccharide stain lactophenol cotton blue was added to Gram stained slides. The dark blue staining is attributable to the presence of capsular polysaccharides and bacterial slime associated with clumps of Gram-negative bacteria.  Since all bacterial cells are glycosylated and concentrate polysaccharides from the media, the majority of cells stain light blue. The contrast between dark and light staining is sufficient to enable a digital image processing thresholding technique to be quantitative with little background noise. Prior to the addition of lactophenol cotton blue, the Gram-stained slides appeared unremarkable, lacking ubiquitous clumps or stained polysaccharides.  Adding lactophenol cotton blue to Gram stained slides is a quick and inexpensive way to screen cell cultures for bacterial slime, clumps and biofilms that are invisible using the Gram stain alone.  The presence of cell clumping provides a possible explanation of the presence of persisters and paradoxical points observed in Virtual Colony Count antimicrobial assays, and suggests a phenotypic resistance mechanism to antimicrobial peptides involving capsular polysaccharides.

13.
F1000Res ; 3: 267, 2014.
Article in English | MEDLINE | ID: mdl-25671086

ABSTRACT

The virtual colony count (VCC) microbiological assay has been utilized for over a decade to measure the antimicrobial activity of peptides such as defensins and LL-37 against biosafety level (BSL)-1 and BSL-2 bacteria including Escherichia coli, Staphylococcus aureus, Bacillus cereus, and Enterobacter aerogenes.  In addition, a modified pipetting technique was presented in a 2011 study of defensin activity against the BSL-3 pathogen Bacillus anthracis.  Both studies were published in the journal Antimicrobial Agents and Chemotherapy.  Here we report that the method can also detect cross-contamination caused by aerosols utilizing the VCC method of data analysis by quantitative growth kinetics (QGK).  The QGK threshold time, or T t, equivalent to the cycle time C t reported in 1996 by Heid et al., precisely identifies when wells were inoculated.

14.
PLoS One ; 8(11): e78937, 2013.
Article in English | MEDLINE | ID: mdl-24236072

ABSTRACT

HNP1 is a human alpha defensin that forms dimers and multimers governed by hydrophobic residues, including Tyr¹6, Ile²°, Leu²5, and Phe²8. Previously, alanine scanning mutagenesis identified each of these residues and other hydrophobic residues as important for function. Here we report further structural and functional studies of residues shown to interact with one another across oligomeric interfaces: I20A-HNP1 and L25A-HNP1, plus the double alanine mutants I20A/L25A-HNP1 and Y16A/F28A-HNP1, and the quadruple alanine mutant Y16A/I20A/L25A/F28A-HNP1. We tested binding to HIV-1 gp120 and HNP1 by surface plasmon resonance, binding to HIV-1 gp41 and HNP1 by fluorescence polarization, inhibition of anthrax lethal factor, and antibacterial activity using the virtual colony count assay. Similar to the previously described single mutant W26A-HNP1, the quadruple mutant displayed the least activity in all functional assays, followed by the double mutant Y16A/F28A-HNP1. The effects of the L25A and I20A single mutations were milder than the double mutant I20A/L25A-HNP1. Crystallographic studies confirmed the correct folding and disulfide pairing, and depicted an array of dimeric and tetrameric structures. These results indicate that side chain hydrophobicity is the critical factor that determines activity at these positions.


Subject(s)
alpha-Defensins/chemistry , Amino Acid Substitution , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Crystallography, X-Ray , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp41/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Mutagenesis, Site-Directed , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , alpha-Defensins/genetics , alpha-Defensins/pharmacology
15.
J Med Chem ; 55(15): 6881-7, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22780881

ABSTRACT

Dengue virus (DENV) infection is a serious public health threat worldwide that demands effective treatment. In the search for potent virus protease inhibitors, several cone snail venoms were screened against serotype 2 DENV NS2B-NS3 protease, and one conotoxin, MrIA, was identified to have inhibitory activity. The inhibitory activity was attributed to a disulfide bond-mediated loop, from which rational optimization was made to improve the potency and stability. An eight-residue cyclic peptide inhibitor was finally obtained with high potency (inhibitory constant 2.2 µM), stability, and cell permeability. This inhibitor can thus serve as a good lead for DENV drug development. In addition, this work highlights the critical effect of peptide cyclization on the potency of oligopeptide inhibitors against DENV protease, which may advance the design of peptide inhibitors for homologous virus proteases.


Subject(s)
Antiviral Agents/chemistry , Conotoxins/chemistry , Dengue Virus/enzymology , Oligopeptides/chemistry , Peptides, Cyclic/chemistry , Serine Endopeptidases , Serine Proteinase Inhibitors/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cell Line , Cell Membrane Permeability , Chlorocebus aethiops , Conotoxins/pharmacology , Cricetinae , Cyclization , Models, Molecular , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacology , Structure-Activity Relationship , Viral Nonstructural Proteins/metabolism
16.
Antimicrob Agents Chemother ; 49(1): 269-75, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15616305

ABSTRACT

We developed a kinetic, 96-well turbidimetric procedure that is capable of testing the antimicrobial properties of six human alpha-defensins concurrently on a single microplate. The defensins were prepared by solid-phase peptide synthesis and tested against gram-positive bacteria (Staphylococcus aureus and Bacillus cereus) and gram-negative bacteria (Enterobacter aerogenes and Escherichia coli). Analysis of the growth curves provided virtual lethal doses (vLDs) equivalent to conventional 50% lethal doses (LD(50)s), LD(90)s, LD(99)s, and LD(99.9)s obtained from colony counts. On the basis of their respective vLD(90)s and vLD(99)s, the relative potencies of human myeloid alpha-defensins against S. aureus were HNP2 > HNP1 > HNP3 > HNP4. In contrast, their relative potencies against E. coli and E. aerogenes were HNP4 > HNP2 > HNP1 = HNP3. HD5 was as effective as HNP2 against S. aureus and as effective as HNP4 against the gram-negative bacteria in our panel. HD6 showed little or no activity against any of the bacteria in our panel, including B. cereus, which was highly susceptible to the other five alpha-defensins. The assay described provides a quantitative, precise, and economical way to study the antimicrobial activities of host-defense peptides. Its use has clarified the relative potencies of human alpha-defensins and raised intriguing questions about the in vivo function(s) of HD6.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , alpha-Defensins/pharmacology , Colony Count, Microbial , Humans , Microbial Sensitivity Tests/methods , Species Specificity , alpha-Defensins/chemical synthesis
17.
J Biol Chem ; 280(52): 43039-47, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16246847

ABSTRACT

Mammalian alpha-defensins, expressed primarily in leukocytes and epithelia, kill a broad range of microbes, constituting one of the first lines of innate immune defense against infection. Nine amino acid residues, including six cysteines, one glycine, and a pair of oppositely charged residues Arg/Glu, are conserved in the otherwise diverse sequences of all known mammalian alpha-defensins. Structural analysis indicates that the two charged residues form a salt bridge, likely stabilizing a protruding loop in the molecule. To investigate the structural and functional roles of the conserved Arg5-Glu13 salt bridge in alpha-defensins, we chemically prepared human neutrophil alpha-defensin 2 (HNP2) and five HNP2 analogs, R5E/E13R, E13Q, E13R, R5T/E13Y, and R14A. In contrast to HNP2 and R14A-HNP2, none of the four salt bridge analogs was capable of folding into a native conformation in the context of isolated defensin domains. However, when covalently attached to the 45-residue pro-HNP2 propeptide, the salt bridge analogs of HNP2 in their pro-forms all folded productively, suggesting that the Arg5-Glu13 salt bridge is not required for correct pro-alpha-defensin folding. When assayed against both Escherichia coli and Staphylococcus aureus, the six alpha-defensins showed bactericidal activity that correlated with the number of net positive charges carried by individual molecules in the panel, irrespective of whether or not the Arg5-Glu13 salt bridge was decimated, suggesting that Arg5 and Glu13 are not functionally conserved. Proteolytic resistance analysis with human neutrophil elastase, one major protease contained in azurophils with HNPs, revealed that destabilization of the salt bridge dramatically accelerated defensin degradation by the enzyme. Thus, we propose that the Arg5-Glu13 salt bridge found in most mammalian alpha-defensins is conserved for defensin in vivo stability.


Subject(s)
Arginine/chemistry , Glutamic Acid/chemistry , alpha-Defensins/chemistry , Alleles , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Conserved Sequence , Cysteine/chemistry , Disulfides/chemistry , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Glycine/chemistry , Humans , Mice , Models, Molecular , Molecular Sequence Data , Neutrophils/metabolism , Oxygen/chemistry , Pancreatic Elastase/chemistry , Peptides/chemistry , Protein Folding , Salts/chemistry , Sequence Homology, Amino Acid , Sodium Chloride/pharmacology , Staphylococcus aureus/metabolism , Time Factors
18.
J Biol Chem ; 280(38): 32921-9, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-15894545

ABSTRACT

Defensins are cationic antimicrobial mini-proteins that play important roles in the innate immune defense against microbial infection. Six invariant Cys residues in each defensin form three structurally indispensable intramolecular disulfide bridges. The only other residue invariant in all known mammalian defensins is a Gly. Structural studies indicate that the invariant Gly residue is located in an atypical, classic-type beta-bulge with the backbone torsion angles (Phi, Psi) disallowed for L-amino acids but permissible for D-enantiomers. We replaced the invariant Gly17 residue in human neutrophil alpha-defensin 2 (HNP2) by L-Ala or one of the D-amino acids Ala, Glu, Phe, Arg, Thr, Val, or Tyr. Although L-Ala17-HNP2 could not be folded, resulting in massive aggregation, all of the D-amino acid-substituted analogs folded with high efficiency. The high resolution x-ray crystal structures of dimeric D-Ala17-HNP2 were determined in three different crystal forms, showing a well preserved beta-bulge identical to those found in other defensins. The seven D-analogs of HNP2 exhibited highly variable bactericidal activity against Gram-positive and Gram-negative test strains, consistent with the premise that interplay between charge and hydrophobicity dictates how amphiphilic defensins kill. Further, the bactericidal activity of these d-amino acid analogs of HNP2 correlated well with their ability to induce leakage from large unilamellar vesicles, supporting membrane permeabilization as the lethal event in microbial killing by HNP2. Our findings identify a conformational prerequisite in the beta-bulge of defensins essential for correct folding and native structure, thereby explaining the molecular basis of the Gly-Xaa-Cys motif conserved in all mammalian defensins.


Subject(s)
Amino Acids/chemistry , Defensins/chemistry , Protein Engineering/methods , Amino Acid Motifs , Amino Acid Sequence , Animals , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Cysteine/chemistry , Disulfides , Dose-Response Relationship, Drug , Glycine/chemistry , Guinea Pigs , Humans , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Neutrophils/chemistry , Oxygen/chemistry , Protein Conformation , Protein Folding , Protein Structure, Secondary , Rabbits , Rats , Spectrometry, Mass, Electrospray Ionization , Static Electricity , Swine , Time Factors , alpha-Defensins/chemistry
19.
Proc Natl Acad Sci U S A ; 101(32): 11587-92, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15280532

ABSTRACT

The HIV-1 matrix protein p17, excised proteolytically from the N terminus of the Gag polyprotein, forms a protective shell attached to the inner surface of the plasma membrane of the virus. During the late stages of the HIV-1 replication cycle, the N-terminally myristoylated p17 domain targets the Gag polyprotein to the host-cell membrane for particle assembly. In the early stages of HIV-1 replication, however, some p17 molecules dissociate from the viral membrane to direct the preintegration complex to the host-cell nucleus. These two opposing targeting functions of p17 require that the protein be capable of reversible membrane interaction. It is postulated that a significant structural change in p17 triggered by proteolytic cleavage of the Gag polyprotein sequesters the N-terminal myristoyl group, resulting in a weaker membrane binding by the matrix protein than the Gag precursor. To test this "myristoyl switch" hypothesis, we obtained highly purified synthetic HIV-1 p17 of 131 amino acid residues and its N-myristoylated form in large quantity. Both forms of p17 were characterized by circular dichroism spectroscopy, protein chemical denaturation, and analytical centrifugal sedimentation. Our results indicate that although N-myristoylation causes no spectroscopically discernible conformational change in p17, it stabilizes the protein by 1 kcal/mol and promotes protein trimerization in solution. These findings support the premise that the myristoyl switch in p17 is triggered not by a structural change associated with proteolysis, but rather by the destabilization of oligomeric structures of membrane-bound p17 in the absence of downstream Gag subdomains.


Subject(s)
Gene Products, gag/chemical synthesis , Myristates/chemical synthesis , Viral Proteins/chemical synthesis , Cell Membrane/metabolism , Dimerization , Gene Products, gag/chemistry , Gene Products, gag/metabolism , HIV Antigens/chemistry , HIV Antigens/metabolism , Protein Binding , Protein Conformation , Protein Denaturation , Spectrum Analysis , Viral Proteins/chemistry , Viral Proteins/metabolism , gag Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL