Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
AIDS Res Hum Retroviruses ; 30(3): 241-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24134315

ABSTRACT

A common route for HIV-1 infection is sexual transmission across colorectal mucosa, which is thought to be 10-2,000 times more vulnerable to infection than that of the female genital tract. Mucosal surfaces are the first line of defense against many pathogens but the antigen-presenting cells (APCs), key regulators of innate immunity and determinants of adaptive immunity, are not well defined in these target tissues. Using immunohistochemistry, dendritic cells expressing Langerin (CD207(+)), a lectin known to bind and internalize HIV-1, were detected in the periphery of colonic glands and sparsely scattered in the submucosa similarly in colorectal mucosa. This cell type, well known in skin, has generally not been reported in colonic/rectal mucosa. Unexpectedly, the largest APC population observed was a macrophage-like population expressing the well-characterized tissue macrophage markers CD68 and CD163. Confocal microscopy of these cells revealed colocalization of CD209 (DC-SIGN), a presumed dendritic cell marker believed to facilitate HIV-1 transmission, but not other dendritic cell markers. These results show evidence of the unconfirmed presence of Langerhans cells in colorectal mucosa and a predominance of macrophage-like APCs that express CD209 (DC-SIGN). These findings define potential target cells in the pathogenesis of HIV-1 transmission, which may have key implications for the study of early transmission events in normal colorectal mucosa, as well as other infectious diseases and primary immune diseases involving the gut.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, CD/analysis , Cell Adhesion Molecules/analysis , Colon/cytology , Dendritic Cells/immunology , Intestinal Mucosa/cytology , Lectins, C-Type/analysis , Macrophages/immunology , Mannose-Binding Lectins/analysis , Receptors, Cell Surface/analysis , Colon/immunology , Dendritic Cells/chemistry , HIV Infections/immunology , HIV Infections/transmission , Humans , Intestinal Mucosa/immunology , Macrophages/chemistry , Male
2.
Biopolymers ; 100(5): 519-26, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23897622

ABSTRACT

The peptide hormone hepcidin is a key regulator of iron homeostasis in vertebrates. Hepcidin acts by binding to ferroportin, the sole known iron exporter, causing it to be internalized and thus trapping iron within the cell. Dysregulation of hepcidin concentrations is associated with a range of iron-related diseases and hepcidin-based therapeutics could be developed as candidate treatments for these diseases. However peptide-based drugs, despite their many advantages, are often limited by their susceptibility to degradation within the body. Here we describe the design, synthesis and characterization of a series of backbone cyclized hepcidin analogues as an approach to produce stable hepcidin-based leads. The cyclic peptides were shown by NMR to be structurally analogous to native hepcidin. Comparison of the stability of hepcidin with one of the cyclic analogues in human serum revealed that 77% of the cyclic peptide but only 18% of linear hepcidin remained after 24 h. The cyclic peptides were tested for their ability to induce internalization of GFP-ferroportin in vitro but were all found to be inactive. This study demonstrates that backbone cyclization of disulfide-rich peptides is a suitable approach for increasing stability. However, careful consideration of a number of factors, including location of important residues and their bioactive conformation, is required to generate biologically active lead molecules.


Subject(s)
Hepcidins , Iron , Animals , Antimicrobial Cationic Peptides/chemistry , Cell Line , Disulfides , Humans , Iron/metabolism , Peptide Hormones , Protein Binding
3.
Blood ; 120(18): 3829-36, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22990014

ABSTRACT

The deficiency of hepcidin, the hormone that controls iron absorption and its tissue distribution, is the cause of iron overload in nearly all forms of hereditary hemochromatosis and in untransfused iron-loading anemias. In a recent study, we reported the development of minihepcidins, small drug-like hepcidin agonists. Here we explore the feasibility of using minihepcidins for the prevention and treatment of iron overload in hepcidin-deficient mice. An optimized minihepcidin (PR65) was developed that had superior potency and duration of action compared with natural hepcidin or other minihepcidins, and favorable cost of synthesis. PR65 was administered by subcutaneous injection daily for 2 weeks to iron-depleted or iron-loaded hepcidin knockout mice. PR65 administration to iron-depleted mice prevented liver iron loading, decreased heart iron levels, and caused the expected iron retention in the spleen and duodenum. At high doses, PR65 treatment also caused anemia because of profound iron restriction. PR65 administration to hepcidin knockout mice with pre-existing iron overload had a more moderate effect and caused partial redistribution of iron from the liver to the spleen. Our study demonstrates that minihepcidins could be beneficial in iron overload disorders either used alone for prevention or possibly as adjunctive therapy with phlebotomy or chelation.


Subject(s)
Antimicrobial Cationic Peptides/agonists , Antimicrobial Cationic Peptides/deficiency , Hemochromatosis/prevention & control , Anemia, Iron-Deficiency/chemically induced , Animals , Antimicrobial Cationic Peptides/pharmacology , Chromatography, High Pressure Liquid , Disease Models, Animal , Hepcidins , Iron Overload/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
J Clin Invest ; 121(12): 4880-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22045566

ABSTRACT

Iron overload is the hallmark of hereditary hemochromatosis and a complication of iron-loading anemias such as ß-thalassemia. Treatment can be burdensome and have significant side effects, and new therapeutic options are needed. Iron overload in hereditary hemochromatosis and ß-thalassemia intermedia is caused by hepcidin deficiency. Although transgenic hepcidin replacement in mouse models of these diseases prevents iron overload or decreases its potential toxicity, natural hepcidin is prohibitively expensive for human application and has unfavorable pharmacologic properties. Here, we report the rational design of hepcidin agonists based on the mutagenesis of hepcidin and the hepcidin-binding region of ferroportin and computer modeling of their docking. We identified specific hydrophobic/aromatic residues required for hepcidin-ferroportin binding and obtained evidence in vitro that a thiol-disulfide interaction between ferroportin C326 and the hepcidin disulfide cage may stabilize binding. Guided by this model, we showed that 7­9 N-terminal amino acids of hepcidin, including a single thiol cysteine, comprised the minimal structure that retained hepcidin activity, as shown by the induction of ferroportin degradation in reporter cells. Further modifications to increase resistance to proteolysis and oral bioavailability yielded minihepcidins that, after parenteral or oral administration to mice, lowered serum iron levels comparably to those after parenteral native hepcidin. Moreover, liver iron concentrations were lower in mice chronically treated with minihepcidins than those in mice treated with solvent alone. Minihepcidins may be useful for the treatment of iron overload disorders.


Subject(s)
Antimicrobial Cationic Peptides/agonists , Iron Overload/drug therapy , Peptide Fragments/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/deficiency , Antimicrobial Cationic Peptides/genetics , Binding Sites , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Computer Simulation , Cysteine/chemistry , Drug Design , Drug Evaluation, Preclinical , Hepcidins , Humans , Hydrophobic and Hydrophilic Interactions , Iron/blood , Liver/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/therapeutic use , Protein Conformation , Protein Interaction Mapping , Structure-Activity Relationship
5.
Chem Biol ; 18(3): 336-43, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21439478

ABSTRACT

The peptide hormone hepcidin is a key homeostatic regulator of iron metabolism and involved in pathological regulation of iron in response to infection, inflammation, hypoxia, and anemia. It acts by binding to the iron exporter ferroportin, causing it to be internalized and degraded; however, little is known about the structure/activity relationships of the interaction of hepcidin with ferroportin. We show that there are key residues in the N-terminal region of hepcidin that influence its interaction with ferroportin, and we explore the structure/function relationships at these positions. A series of hepcidin mutants in which disulfide bonds were replaced with diselenide bonds showed no change in activity compared to native hepcidin. These results identify important constraints for the development of hepcidin congeners for the treatment of hereditary iron overload.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Iron/metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides/chemical synthesis , Cation Transport Proteins/chemistry , Cation Transport Proteins/metabolism , Disulfides/chemistry , Hepcidins , Humans , Iron Overload/therapy , Molecular Sequence Data , Protein Structure, Tertiary , Selenium/chemistry , Structure-Activity Relationship
6.
Blood ; 114(2): 437-43, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19383972

ABSTRACT

The interaction between the hormone hepcidin and the iron exporter ferroportin (Fpn) regulates plasma iron concentrations. Hepcidin binds to Fpn and induces its internalization and degradation, resulting in decreased iron efflux from cells into plasma. Fpn mutations in N144, Y64N, and C326 residue cause autosomal dominant disease with parenchymal iron overload, apparently due to the resistance of mutant Fpn to hepcidin-mediated internalization. To define the mechanism of resistance, we generated human Fpn constructs bearing the pathogenic mutations. The mutants localized to the cell surface and exported iron normally, but were partially or completely resistant to hepcidin-mediated internalization and continued to export iron despite the presence of hepcidin. The primary defect with exofacial C326 substitutions was the loss of hepcidin binding, which resulted in the most severe phenotype. The thiol form of C326 was essential for interaction with hepcidin, suggesting that C326-SH homology is located in or near the binding site of hepcidin. In contrast, N144 and Y64 residues were not required for hepcidin binding, but their mutations impaired the subsequent internalization of the ligand-receptor complex. Our observations explain why the mutations in C326 Fpn residue produce a severe form of hemochromatosis with iron overload at an early age.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Hemochromatosis/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Hemochromatosis/genetics , Hepcidins , Humans , Iron/metabolism , Ligands , Mutation/genetics , Protein Binding , Sulfhydryl Compounds/metabolism
7.
Blood ; 107(1): 328-33, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16141345

ABSTRACT

Hepcidin is the principal iron-regulatory hormone. It acts by binding to the iron exporter ferroportin, inducing its internalization and degradation, thereby blocking cellular iron efflux. The bioactive 25 amino acid (aa) peptide has a hairpin structure stabilized by 4 disulfide bonds. We synthesized a series of hepcidin derivatives and determined their bioactivity in a cell line expressing ferroportin-GFP fusion protein, by measuring the degradation of ferroportin-GFP and the accumulation of ferritin after peptide treatment. Bioactivity was also assayed in mice by the induction of hypoferremia. Serial deletion of N-terminal amino acids caused progressive decrease in activity which was completely lost when 5 N-terminal aa's were deleted. Synthetic 3-aa and 6-aa N-terminal peptides alone, however, did not internalize ferroportin and did not interfere with ferroportin internalization by native hepcidin. Deletion of 2 C-terminal aa's did not affect peptide activity. Removal of individual disulfide bonds by pairwise substitution of cysteines with alanines also did not affect peptide activity in vitro. However, these peptides were less active in vivo, likely because of their decreased stability in circulation. G71D and K83R, substitutions previously described in humans, did not affect hepcidin activity. Apart from the essential nature of the N-terminus, hepcidin structure appears permissive for mutations.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Cation Transport Proteins/metabolism , Peptides/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antimicrobial Cationic Peptides/genetics , Binding Sites , Cation Transport Proteins/genetics , Cell Line , Disulfides , Ferritins/metabolism , Hepcidins , Mice , Mice, Inbred C57BL , Mutation , Peptides/chemical synthesis , Sequence Deletion , Structure-Activity Relationship , Transfection
8.
Infect Immun ; 73(8): 4823-33, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16040996

ABSTRACT

Defensins are key participants in mucosal innate defense. The varied antimicrobial activity and differential distribution of defensins at mucosal sites indicate that peptide repertoires are tailored to site-specific innate defense requirements. Nonetheless, few studies have investigated changes in peptide profiles and function after in vivo pathogen challenge. Here, we determined defensin profiles in urethral secretions of healthy men and men with Chlamydia trachomatis- and Neisseria gonorrhoeae-mediated urethritis by immunoblotting for the epithelial defensins HBD1, HBD2, and HD5 and the neutrophil defensins HNP1 to -3 (HNP1-3). HBD1 was not detectable in secretions, and HBD2 was only induced in a small proportion of the urethritis patients; however, HD5 and HNP1-3 were increased in C. trachomatis infection and significantly elevated in N. gonorrhoeae infection. When HNP1-3 levels were low, HD5 appeared mostly as the propeptide; however, when HNP1-3 levels were >10 microg/ml, HD5 was proteolytically processed, suggesting neutrophil proteases might contribute to HD5 processing. HD5 and HNP1-3 were bactericidal against C. trachomatis and N. gonorrhoeae, but HD5 activity was dependent upon N-terminal processing of the peptide. In vitro proteolysis of proHD5 by neutrophil proteases and analysis of urethral secretions by surface-enhanced laser desorption ionization substantiated that neutrophils contribute the key convertases for proHD5 in the urethra during these infections. This contrasts with the small intestine, where Paneth cells secrete both proHD5 and its processing enzyme, trypsin. In conclusion, we describe a unique defensin expression repertoire in response to inflammatory sexually transmitted infections and a novel host defense mechanism wherein epithelial cells collaborate with neutrophils to establish an antimicrobial barrier during infection.


Subject(s)
Chlamydia Infections/metabolism , Defensins/metabolism , Epithelial Cells/metabolism , Gonorrhea/metabolism , Neutrophils/metabolism , Urethritis/metabolism , Chlamydia trachomatis , Humans , Immunoblotting , Male , Neisseria gonorrhoeae , Neutrophils/enzymology , Peptide Hydrolases/metabolism , Urethra/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...