Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 295(7): 1781-1791, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31819010

ABSTRACT

Iron is an essential nutrient for all living organisms. To acquire iron, many pathogens have developed elaborate systems to steal it from their hosts. The iron acquisition system in the opportunistic pathogen Staphylococcus aureus comprises nine proteins, called iron-regulated surface determinants (Isds). The Isd components enable S. aureus to extract heme from hemoglobin (Hb), transport it into the bacterial cytoplasm, and ultimately release iron from the porphyrin ring. IsdB and IsdH act as hemoglobin receptors and are known to actively extract heme from extracellular Hb. To limit microbial pathogenicity during infection, host organisms attempt to restrict the availability of nutrient metals at the host-pathogen interface. The human acute phase protein haptoglobin (Hp) protects the host from oxidative damage by clearing hemoglobin that has leaked from red blood cells and also restricts the availability of extracellular Hb-bound iron to invading pathogens. To investigate whether Hp serves an additional role in nutritional immunity through a direct inhibition of IsdH-mediated iron acquisition, here we measured heme extraction from the Hp-Hb complex by UV-visible spectroscopy and determined the crystal structure of the Hp-Hb-IsdH complex at 2.9 Å resolution. We found that Hp strongly inhibits IsdH-mediated heme extraction and that Hp binding prevents local unfolding of the Hb heme pocket, leaving IsdH unable to wrest the heme from Hb. Furthermore, we noted that the Hp-Hb binding appears to trap IsdH in an initial state before heme transfer. Our findings provide insights into Hp-mediated IsdH inhibition and the dynamics of IsdH-mediated heme extraction.


Subject(s)
Antigens, Bacterial/chemistry , Haptoglobins/chemistry , Host-Pathogen Interactions/genetics , Receptors, Cell Surface/chemistry , Staphylococcal Infections/genetics , Crystallography, X-Ray , Erythrocytes/metabolism , Erythrocytes/microbiology , Haptoglobins/genetics , Haptoglobins/ultrastructure , Heme/chemistry , Heme/genetics , Hemoglobins/chemistry , Hemoglobins/genetics , Humans , Iron/chemistry , Iron/metabolism , Protein Binding/genetics , Protein Conformation , Receptors, Cell Surface/antagonists & inhibitors , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity
2.
J Biol Chem ; 290(36): 21915-24, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26195635

ABSTRACT

Stanniocalcin-1 (STC1) is a disulfide-bound homodimeric glycoprotein, first identified as a hypocalcemic hormone important for maintaining calcium homeostasis in teleost fish. STC1 was later found to be widely expressed in mammals, although it is not believed to function in systemic calcium regulation in these species. Several physiological functions of STC1 have been reported, although many molecular details are still lacking. We here demonstrate that STC1 is an inhibitor of the metzincin metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A), which modulates insulin-like growth factor (IGF) signaling through proteolytic cleavage of IGF-binding proteins (IGFBPs). STC1 potently (Ki = 68 pm) inhibits PAPP-A cleavage of IGFBP-4, and we show in a cell-based assay that STC1 effectively antagonizes PAPP-A-mediated type 1 IGF receptor (IGF1R) phosphorylation. It has recently been found that the homologous STC2 inhibits PAPP-A proteolytic activity, and that this depends on the formation of a covalent complex between the inhibitor and the proteinase, mediated by Cys-120 of STC2. We find that STC1 is unable to bind covalently to PAPP-A, in agreement with the absence of a corresponding cysteine residue. It rather binds to PAPP-A with high affinity (KD = 75 pm). We further demonstrate that both STC1 and STC2 show inhibitory activity toward PAPP-A2, but not selected serine proteinases and metalloproteinases. We therefore conclude that the STCs are proteinase inhibitors, probably restricted in specificity to the pappalysin family of metzincin metalloproteinases. Our data are the first to identify STC1 as a proteinase inhibitor, suggesting a previously unrecognized function of STC1 in the IGF system.


Subject(s)
Glycoproteins/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Blotting, Western , Glycoproteins/genetics , Glycoproteins/pharmacology , HEK293 Cells , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Phosphorylation/drug effects , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/genetics , Proteolysis/drug effects , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
3.
J Biol Chem ; 290(6): 3430-9, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25533459

ABSTRACT

Mammalian stanniocalcin-2 (STC2) is a secreted polypeptide widely expressed in developing and adult tissues. However, although transgenic expression in mice is known to cause severe dwarfism, and targeted deletion of STC2 causes increased postnatal growth, its precise biological role is still unknown. We found that STC2 potently inhibits the proteolytic activity of the growth-promoting metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A). Proteolytic inhibition requires covalent binding of STC2 to PAPP-A and is mediated by a disulfide bond, which involves Cys-120 of STC2. Binding of STC2 prevents PAPP-A cleavage of insulin-like growth factor-binding protein (IGFBP)-4 and hence release within tissues of bioactive IGF, required for normal growth. Concordantly, we show that STC2 efficiently inhibits PAPP-A-mediated IGF receptor signaling in vitro and that transgenic mice expressing a mutated variant of STC2, STC2(C120A), which is unable to inhibit PAPP-A, grow like wild-type mice. Our work identifies STC2 as a novel proteinase inhibitor and a previously unrecognized extracellular component of the IGF system.


Subject(s)
Glycoproteins/metabolism , Growth/genetics , Pregnancy-Associated Plasma Protein-A/metabolism , Proteolysis , Amino Acid Sequence , Animals , Cells, Cultured , Cysteine/chemistry , Cysteine/genetics , Female , Glycoproteins/chemistry , Glycoproteins/genetics , HEK293 Cells , Humans , Insulin-Like Growth Factor Binding Protein 4/metabolism , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Male , Mice , Molecular Sequence Data , Mutation , Protein Binding , Receptor, IGF Type 1/metabolism
4.
Oncotarget ; 5(4): 1014-25, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24572990

ABSTRACT

The insulin-like growth factor (IGF) signaling pathway is involved in certain human cancers, and the feasibility of directly targeting the IGF receptor has been actively investigated. However, recent evidence from clinical trials suggests that this approach can be problematic. We have developed an alternative strategy to indirectly inhibit the IGF signaling by targeting the metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A). PAPP-A associated with the cell surface cleaves IGF binding protein-4 (IGFBP-4), when IGF is bound to IGFBP-4, and thereby increases IGF bioavailability for receptor activation in an autocrine/paracrine manner. We hypothesized that inhibition of PAPP-A would suppress excessive local IGF signaling in tissues where this is caused by increased PAPP-A proteolytic activity. To test this hypothesis, we developed an inhibitory monoclonal antibody, mAb 1/41, which targets a unique substrate-binding exosite of PAPP-A. This inhibitor selectively and specifically inhibits proteolytic cleavage of IGFBP-4 with an inhibitory constant (Ki) of 135 pM. In addition, it inhibited intracellular signaling of the IGF receptor (AKT phosphorylation) in monolayers of A549 cells, an IGF-responsive lung cancer-derived cell line found to express high levels of PAPP-A. We further showed that mAb 1/41 is effective towards PAPP-A bound to cell surfaces, and that it is capable of inhibiting PAPP-A activity in vivo. Using a murine xenograft model of A549 cells, we demonstrated that mAb 1/41 administered intraperitoneally significantly inhibited tumor growth. Analysis of xenograft tumor tissue recovered from treated mice showed penetration of mAb 1/41, reduced IGFBP-4 proteolysis, and reduced AKT phosphorylation. Our study provides proof of concept that IGF signaling can be selectively reduced by targeting a regulatory proteinase that functions extracellularly, upstream of the IGF receptor. PAPP-A targeting thus represents an alternative therapeutic strategy for inhibiting IGF receptor signaling.


Subject(s)
Antibodies, Monoclonal/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Receptors, Somatomedin/metabolism , Animals , Antibodies, Monoclonal/immunology , Disease Models, Animal , Female , HEK293 Cells , Heterografts , Humans , Insulin-Like Growth Factor I/metabolism , Lung Neoplasms/enzymology , Male , Mice , Mice, Knockout , Molecular Targeted Therapy , Pregnancy , Pregnancy-Associated Plasma Protein-A/immunology , Pregnancy-Associated Plasma Protein-A/pharmacology , Signal Transduction , Transfection , Xenograft Model Antitumor Assays
5.
J Immunol Methods ; 404: 33-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24333852

ABSTRACT

The metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A), is increasingly recognized as a modulator of insulin-like growth factor (IGF) signaling; it cleaves IGF binding proteins causing the release of bioactive IGF. Accumulating evidence supports an important role of PAPP-A in both normal physiology and under different pathological conditions. However, antibodies for the detection of PAPP-A in non-human tissues have been lacking, although needed for use with several animal models which are currently being developed. To develop a monoclonal antibody suitable for the immunohistochemical detection of PAPP-A, we therefore selected a phage-derived scFv antibody, PAC1, specifically recognizing an epitope of PAPP-A, which is highly conserved between multiple animal species. We first converted this antibody into bivalent IgG, and verified its ability to recognize PAPP-A in sections of formalin-fixed and paraffin-embedded tissue. For increased sensitivity, affinity maturation to sub-nanomolar affinity was then carried out. The resulting recombinant antibody, PAC1-D8-mIgG2a, detects PAPP-A specifically and sensitively in human tissue. In addition, this antibody allows detection of PAPP-A in non-human species. We demonstrate its usefulness for the visualization of PAPP-A in murine and porcine tissues.


Subject(s)
Immunoglobulin G/biosynthesis , Immunohistochemistry/methods , Pregnancy-Associated Plasma Protein-A/analysis , Single-Chain Antibodies/biosynthesis , Amino Acid Sequence , Animals , Antibody Affinity , Antibody Specificity , Conserved Sequence , Epitopes/chemistry , Epitopes/immunology , Female , Formaldehyde , Humans , Immunoglobulin G/immunology , Mice , Molecular Sequence Data , Paraffin Embedding , Pregnancy , Pregnancy-Associated Plasma Protein-A/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Sequence Alignment , Single-Chain Antibodies/immunology , Swine , Tissue Fixation
6.
J Endocrinol ; 219(1): 51-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23881937

ABSTRACT

We investigated pregnancy-associated plasma protein-A (PAPP-A) in diabetic nephropathy. Normal human kidney showed specific staining for PAPP-A in glomeruli, and this staining was markedly increased in diabetic kidney. To assess the possible contribution of PAPP-A in the development of diabetic nephropathy, we induced diabetes with streptozotocin in 14-month-old WT and Papp-A knockout (KO) mice. Renal histopathology was evaluated after 4 months of stable hyperglycemia. Kidneys from diabetic WT mice showed multiple abnormalities including thickening of Bowman's capsule (100% of mice), increased glomerular size (80% of mice), tubule dilation (80% of mice), and mononuclear cell infiltration (90% of mice). Kidneys of age-matched non-diabetic WT mice had similar evidence of tubule dilation and mononuclear cell infiltration to those of diabetic WT mice, indicating that these changes were predominantly age-related. However, thickened Bowman's capsule and increased glomerular size appeared specific for the experimental diabetes. Kidneys from diabetic Papp-A KO mice had significantly reduced or no evidence of changes in Bowman's capsule thickening and glomerular size. There was also a shift to larger mesangial area and increased macrophage staining in diabetic WT mice compared with Papp-A KO mice. In summary, elevated PAPP-A expression in glomeruli is associated with diabetic nephropathy in humans and absence of PAPP-A is associated with resistance to the development of indicators of diabetic nephropathy in mice. These data suggest PAPP-A as a potential therapeutic target for diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/pathology , Kidney/pathology , Pregnancy-Associated Plasma Protein-A/deficiency , Aging/pathology , Animals , Bowman Capsule/pathology , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/etiology , Diabetic Nephropathies/physiopathology , Female , Glomerular Mesangium/pathology , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Pregnancy
7.
J Biol Chem ; 283(24): 16772-80, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18434323

ABSTRACT

The metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) cleaves both insulin-like growth factor (IGF)-binding protein 4 (IGFBP-4) and -5 at a single site in their central domain causing the release of bioactive IGF. Inhibition of IGF signaling is relevant in human disease, and several drugs in development target the IGF receptor. However, inhibition of PAPP-A activity may be a valuable alternative. We have generated monoclonal phage-derived single chain fragment variable (scFv) antibodies which selectively inhibit the cleavage of IGFBP-4 by PAPP-A, relevant under conditions where cleavage of IGFBP-4 represents the final step in the delivery of IGF to the IGF receptor. None of the antibodies inhibited the homologous proteinase PAPP-A2, which allowed mapping of antibody binding by means of chimeras between PAPP-A and PAPP-A2 to the C-terminal Lin12-Notch repeat module, separated from the proteolytic domain by almost 1000 amino acids. Hence, the antibodies define a substrate binding exosite that can be targeted for the selective inhibition of PAPP-A proteolytic activity against IGFBP-4. In addition, we show that the Lin12-Notch repeat module reversibly binds a calcium ion and that bound calcium is required for antibody binding, providing a strategy for the further development of selective inhibitory compounds. To our knowledge these data represent the first example of differential inhibition of cleavage of natural proteinase substrates by exosite targeting. Generally, exosite inhibitors are less likely to affect the activity of related proteolytic enzymes with similar active site environments. In the case of PAPP-A, selective inhibition of IGFBP-4 cleavage by interference with exosite binding is a further advantage, as the activity against other known or unknown PAPP-A substrates, whose cleavage may not depend on binding to the same exosite, is not targeted.


Subject(s)
Pregnancy-Associated Plasma Protein-A/metabolism , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Cell Line , Chickens , Humans , Insulin-Like Growth Factor Binding Protein 4/chemistry , Insulin-Like Growth Factor I/metabolism , Models, Genetic , Peptide Library , Pregnancy-Associated Plasma Protein-A/chemistry , Protein Binding , Protein Structure, Tertiary , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...