Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(8): e71159, 2013.
Article in English | MEDLINE | ID: mdl-24023608

ABSTRACT

Ovarian cancer is the most lethal gynaecological cancer and is often diagnosed in late stage, often as the result of the unavailability of sufficiently sensitive biomarkers for early detection, tumour progression and tumour-associated inflammation. Glycosylation is the most common posttranslational modification of proteins; it is altered in cancer and therefore is a potential source of biomarkers. We investigated the quantitative and qualitative effects of anti-inflammatory (acetylsalicylic acid) and pro-inflammatory (thioglycolate and chlorite-oxidized oxyamylose) drugs on glycosylation in mouse cancer serum. A significant increase in sialylation and branching of glycans in mice treated with an inflammation-inducing compound was observed. Moreover, the increases in sialylation correlated with increased tumour sizes. Increases in sialylation and branching were consistent with increased expression of sialyltransferases and the branching enzyme MGAT5. Because the sialyltransferases are highly conserved among species, the described changes in the ovarian cancer mouse model are relevant to humans and serum N-glycome analysis for monitoring disease treatment and progression might be a useful biomarker.


Subject(s)
Disease Progression , Glycoproteins/blood , Inflammation/blood , Inflammation/pathology , N-Acetylneuraminic Acid/metabolism , Ovarian Neoplasms/blood , Ovarian Neoplasms/pathology , Amylose/administration & dosage , Amylose/analogs & derivatives , Amylose/pharmacology , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Glycoproteins/chemistry , Glycosylation , Humans , Inflammation/complications , Mice , Molecular Weight , Neuraminic Acids/metabolism , Ovarian Neoplasms/complications , Ovarian Neoplasms/immunology , Sialyltransferases/metabolism , Thioglycolates/administration & dosage , Thioglycolates/pharmacology , Tumor Burden/drug effects
2.
Int J Cancer ; 131(4): E425-36, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-21953247

ABSTRACT

Chlorite-oxidized oxyamylose (COAM), a glycosaminoglycan mimetic and potent antiviral agent, provided significant growth reduction of syngeneic murine B16-F1 melanoma tumors. A single early dose (100 µg, into the site of tumor cell inoculation) was sufficient to establish a persistent effect over 17 days (resected tumor volume of 78.3 mm(3) in COAM-treated mice compared to 755.2 mm(3) in the control cohort, i.e., 89.6% reduction of tumor volumes). COAM was a much better antitumoral agent than the polyanionic glycosaminoglycan heparin. COAM retained its antitumoral effect in lymphopenic mice, reinforcing the idea of myeloid cell involvement. Massive recruitment of myeloid cells into dermal air pouches in response to COAM and their increased presence in early-treated tumors indicated that mainly CD11b(+) GR-1(+) myeloid cells were attracted by COAM to exert antitumoral effects. Leukocyte chemotaxis was mediated by the chemokine system through the induction in B16-F1 cells of mouse granulocyte chemotactic protein-2/CXCL6 upon COAM treatment. Thus, COAM constitutes a novel tool to study the role of innate immune cells in the initial stages of tumor development and an example that innate immunostimulating glycosaminoglycan mimicry may be exploited therapeutically.


Subject(s)
Amylose/analogs & derivatives , Chemokines/biosynthesis , Glycosaminoglycans/metabolism , Melanoma, Experimental/pathology , Molecular Mimicry , Amylose/pharmacology , Animals , Cell Division/drug effects , Flow Cytometry , Fluorescent Dyes , Male , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Real-Time Polymerase Chain Reaction
3.
Thromb Haemost ; 104(1): 165-71, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20431850

ABSTRACT

This study was performed to follow up on the observation that gelatinase A (MMP-2) deficiency impairs adipose tissue development in mice. The aim was to evaluate the role of its functional homologue gelatinase B (MMP-9) in adipose tissue growth. MMP-9 antigen levels were determined in lean and in obese women before and after weight loss. MMP-9-deficient mice and wild-type littermates (genetic background 50% 129sv : 50% CDI or 99.975% C57Bl/6, ten generations backcrossed into C57Bl/6 background) were kept on a high-fat diet (HFD) for 15 weeks. Subcutaneous and gonadal fat pads were analysed in terms of weight and size/density of adipocytes and blood vessels. Obese women had higher MMP-9 serum levels than lean controls (383 +/- 29 vs. 304 +/- 27 ng/ml, p = 0.02); after weight reduction MMP-9 levels dropped to 334 +/- 17 ng/ml (p = 0.1 vs. obese). However, MMP-9-deficient and littermate wild-type mice kept on HFD were indistinguishable in terms of body and fat weight. No effect of MMP-9 deficiency was observed on size or density of adipocytes or blood vessels in subcutaneous or gonadal fat depots. Similar observations were made when mice were kept on normal chow. In conclusion, in lean and obese women, body mass index correlates positively with MMP-9 serum levels (p < 0.0001). However, MMP-9 does not seem to play a major role in adipose tissue development in murine models of diet-induced obesity.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Obesity/metabolism , Subcutaneous Fat/cytology , Adult , Animals , Body Mass Index , Cell Differentiation/genetics , Feeding Behavior , Female , Humans , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/pathology , Obesity/physiopathology , Subcutaneous Fat/embryology , Subcutaneous Fat/growth & development
4.
Electrophoresis ; 30(13): 2366-77, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19621364

ABSTRACT

A platform for profiling of multiple proteolytic activities acting on one specific substrate, based on the use of a 96-channel capillary DNA sequencer with CE-LIF of labeled substrate peptides and reaction products is introduced. The approach consists of synthesis of a substrate peptide of interest, fluorescent labeling of the substrate, either aminoterminally by chemical coupling, or carboxyterminally by transglutaminase reaction, proteolysis by a biological mixture of proteases in the absence or presence of protease inhibitors, multi-channel analysis of substrate and reaction products, and data collection and processing. Intact substrate and reaction products, even when varying by only one amino acid, can be relatively semi-quantified in a high-throughput manner, yielding information on proteases acting in complex biological mixtures and without prepurification. Monitoring, classification and inhibition of multiple proteolytic activities are demonstrated on a model substrate, the aminoterminus of the mouse granulocyte chemotactic protein-2. In view of extensive processing of chemokines into various natural forms with different specific biological activities, and of the fragmentary knowledge of processing proteases, examples of processing by neutrophil degranulate, tumor cell culture fluids and plasma are provided. An example of selection and comparison of inhibitory mAbs illustrates that the platform is suitable for inhibitor screening. Whereas classical degradomics technologies analyze the substrate repertoire of one specific protease, here the complementary concept, namely the study of all proteases acting, in a biological context, on one specific substrate, is developed and tuned to identify key proteases and protease inhibitors for the processing of any biological substrate of interest.


Subject(s)
Electrophoresis, Capillary/methods , Fluorescent Dyes/chemistry , Microscopy, Confocal/methods , Peptide Hydrolases/chemistry , Peptides/analysis , Proteomics/methods , Animals , Antibodies, Monoclonal/pharmacology , Chemokine CXCL6/metabolism , Fluorescent Dyes/metabolism , Matrix Metalloproteinase Inhibitors , Mice , Peptide Hydrolases/blood , Peptide Hydrolases/metabolism , Peptide Library , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sensitivity and Specificity , Transglutaminases/metabolism
5.
Immunology ; 128(1 Suppl): e262-74, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19175797

ABSTRACT

Matrix metalloproteinase-9 (MMP-9)/gelatinase B plays an important role in neutrophil infiltration during inflammation and cyclooxygenases (COX-1 and COX-2) and their products are important regulators of inflammation. Recently, we reported that a genetic lack of MMP-9 impairs neutrophil infiltration during early zymosan-induced peritonitis but at later stages (> 24 hr) neutrophils persist in the peritoneal cavity. Here we show that this is the result of impaired apoptosis of MMP-9(-/-)-derived leucocytes. As enhanced COX-1 expression was reported in MMP-9(-/-) mice, we evaluated the hypothesis that altered COX expression induced the above phenomenon as COX-dependent prostaglandins can act either anti-apoptotically (PGE(2)) or pro-apoptotically (PGD(2)). The current data demonstrate that messenger RNA and protein expression of both COX isoforms and their activities are increased in MMP-9(-/-) mice during late peritonitis. Application of selective COX inhibitors revealed enhanced COX-1-dependent PGE(2) production and impaired COX-2-dependent PGD(2) synthesis in MMP-9(-/-) mice. Most importantly, inhibition of COX-1 abolished prolonged neutrophil accumulation in the peritoneal cavity of MMP-9(-/-) mice and increased apoptosis of inflammatory leucocytes. Similarly, weaker apoptosis of MMP-9(-/-) bone marrow neutrophils treated in vitro with zymosan was reversed by COX-1 inhibition. In conclusion, enhanced COX-1 expression is responsible for persistent neutrophil presence in the peritoneum of MMP-9(-/-) mice because of increased synthesis of anti-apoptotic PGE(2). In non-transgenic mice, however, inflammatory leucocytes die apoptotically in the late stages of peritonitis as a result of COX-2-dependent PGD(2) activity. Overall, we show a dependence of COX expression on the presence of MMP-9.


Subject(s)
Apoptosis/immunology , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/biosynthesis , Neutrophil Infiltration , Neutrophils/immunology , Peritonitis/immunology , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/analysis , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/enzymology , Peritonitis/chemically induced , Peritonitis/enzymology , Prostaglandin D2/analysis , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Thiophenes/administration & dosage , Thiophenes/pharmacology , Zymosan/pharmacology
6.
J Leukoc Biol ; 85(3): 374-81, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19088179

ABSTRACT

Extracellular proteolysis of basement membranes and matrix is required for leukocyte diapedesis and migration to the inflammatory focus. Neutrophil elastase (NE) and matrix metalloproteinases (MMPs) are among the enzymes involved in these processes, as shown in mice genetically deprived of such enzymes. However, studies with MMP-9(-/-) mice revealed that albeit neutrophil influx is impaired initially in these animals versus controls, neutrophilia is subsequently augmented during later stages of zymosan peritonitis. MMP-9 as a MMP and NE as a serine protease belong to different enzyme classes. As MMP-9 and NE are produced by neutrophils and have similar biological effects on matrix remodeling, it was evaluated whether enhanced NE activity might compensate for the lack of MMP-9. In genetically uncompromised mice, two waves of NE expression and activity during zymosan peritonitis were observed in inflammatory neutrophils and macrophages at the time of influx of the respective cell populations into the peritoneum. Additionally, NE expression was associated with the activity of resident peritoneal mast cells and macrophages, as their depletion reduced NE activity. Most importantly, the NE mRNA and protein expression and activity were enhanced significantly in MMP-9(-/-) mice during late stages of zymosan peritonitis. In addition, the application of a selective NE inhibitor restrained enhanced neutrophil accumulation significantly. In conclusion, during acute peritoneal inflammation, NE expression and activity increase gradually, facilitating leukocyte influx. Moreover, increased NE activity might compensate for a genetic lack of MMP-9 (as detected in MMP-9(-/-) mice), resulting in delayed accumulation of neutrophils during late zymosan peritonitis.


Subject(s)
Chemotaxis, Leukocyte/immunology , Leukocyte Elastase/metabolism , Matrix Metalloproteinase 9/deficiency , Peritonitis/immunology , Animals , Disease Models, Animal , Inflammation/immunology , Leukocyte Elastase/analysis , Leukocyte Elastase/genetics , Macrophages , Mice , Mice, Knockout , Neutrophils , RNA, Messenger/analysis , Up-Regulation
7.
J Leukoc Biol ; 81(4): 870-92, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17185359

ABSTRACT

The heme-binding hemopexin consists of two, four-bladed propeller domains connected by a linker region. Hemopexin domains are found in different species on the phylogenetic tree and in the human species represented in hemopexin, matrix metalloproteinases (MMPs), vitronectin, and products of the proteoglycan 4 gene. Hemopexin and hemopexin domains of human proteins fulfill functions in activation of MMPs, inhibition of MMPs, dimerization, binding of substrates or ligands, cleavage of substrates, and endocytosis by low-density lipoprotein receptor-related protein-1 (LRP-1; CD91) and LRP-2 (megalin, GP330). Insights into the structures and functions of hemopexin (domains) form the basis for positive or negative interference with the formation of molecular complexes and hence, might be exploited therapeutically in inflammation, cancer, and wound healing.


Subject(s)
Hemopexin/chemistry , Matrix Metalloproteinases/chemistry , Amino Acid Sequence , Dimerization , Enzyme Activation , Hemopexin/metabolism , Hemopexin/physiology , Humans , Ligands , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/physiology , Models, Biological , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Structure-Activity Relationship
8.
Biochim Biophys Acta ; 1770(2): 178-86, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17137715

ABSTRACT

Gelatinase B/matrix metalloproteinase-9 (MMP-9) is a multidomain enzyme functioning in acute and chronic inflammatory and neoplastic diseases. It belongs to a family of more than 20 related zinc proteinases. Therefore, the discovery and the definition of the action mechanism of selective MMP inhibitors form the basis for future therapeutics. The monoclonal antibody REGA-3G12 is a most selective inhibitor of human gelatinase B. REGA-3G12 was found to recognize the aminoterminal part and not the carboxyterminal O-glycosylated and hemopexin protein domains. A variant of gelatinase B, lacking the two carboxyterminal domains, was expressed in insect cells and fragmented with purified proteinases. The fragments were probed by one- and two-dimensional Western blot and immunoprecipitation experiments with REGA-3G12 to map the interactions between the antibody and the enzyme. The interaction unit was identified by Edman degradation analysis as the glycosylated segment from Trp(116) to Lys(214) of gelatinase B. The sequence of this segment was analysed by hydrophobicity/hydrophilicity, accessibility and flexibility profiling. Four hydrophilic peptides were chemically synthesized and used in binding and competition assays. The peptide Gly(171)-Leu(187) in molar excess inhibited partially the binding of MMP-9 to REGA-3G12 and thus refines the structure of the conformational binding site. These results define part of the catalytic domain of gelatinase B/MMP-9, and not the zinc-binding or fibronectin domains, as target for the development of selective inhibitors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Matrix Metalloproteinase Inhibitors , Amino Acid Sequence , Animals , Binding Sites , Catalytic Domain , Cloning, Molecular , DNA, Complementary/genetics , Enzyme Inhibitors/pharmacology , Fibronectins/chemistry , Fibronectins/metabolism , Genetic Variation , Humans , Insecta , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , Molecular Sequence Data , Peptide Fragments/chemistry , Protease Inhibitors/pharmacology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Zinc/metabolism
9.
J Biol Chem ; 281(27): 18626-37, 2006 Jul 07.
Article in English | MEDLINE | ID: mdl-16672230

ABSTRACT

Gelatinase B/matrix metalloproteinase-9 (MMP-9), a key regulator and effector of immunity, contains a C-terminal hemopexin domain preceded by a unique linker sequence of approximately 64 amino acid residues. This linker sequence is demonstrated to be an extensively O-glycosylated (OG) domain with a compact three-dimensional structure. The OG and hemopexin domains have no influence on the cleavage efficiency of MMP-9 substrates. In contrast, the hemopexin domain contains a binding site for the cargo receptor low density lipoprotein receptor-related protein-1 (LRP-1). Furthermore, megalin/LRP-2 is identified as a new functional receptor for the hemopexin domain of MMP-9, able to mediate the endocytosis and catabolism of the enzyme. The OG domain is required to correctly orient the hemopexin domain for inhibition by TIMP-1 and internalization by LRP-1 and megalin. Therefore, the OG and hemopexin domains down-regulate the bioavailability of active MMP-9 and the interactions with the cargo receptors are proposed to be the original function of hemopexin domains in MMPs.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/metabolism , Binding Sites , Catalytic Domain , Down-Regulation , Glycosylation , Hemopexin/chemistry , Humans , Matrix Metalloproteinase 9/genetics , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Tissue Inhibitor of Metalloproteinase-1/metabolism
10.
Biochim Biophys Acta ; 1760(4): 610-5, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16439062

ABSTRACT

The role of extracellular proteolysis in innate and adaptive immunity and the interplay between cytokines, chemokines and proteinases are gradually becoming recognized as critical factors in autoimmune processes. Many of the involved proteinases, including those of the plasminogen activator and matrix metalloproteinase cascades, and also several cytokines and chemokines, are glycoproteins. The stability, interactions with inhibitors or receptors, and activities of these molecules are fine-controlled by glycosylation. We studied gelatinase B or matrix metalloproteinase-9 (MMP-9) as a glycosylated enzyme involved in autoimmunity. In the joints of rheumatoid arthritis patients, CXC chemokines, such as interleukin-8/CXCL8, recruit and activate neutrophils to secrete prestored neutrophil collagenase/MMP-8 and gelatinase B/MMP-9. Gelatinase B potentiates interleukin-8 at least tenfold and thus enhances neutrophil and lymphocyte influxes to the joints. When cartilage collagen type II is cleaved at a unique site by one of several collagenases (MMP-1, MMP-8 or MMP-13), it becomes a substrate of gelatinase B. Human gelatinase B cleaves the resulting two large collagen fragments into at least 33 peptides of which two have been shown to be immunodominant, i.e., to elicit activation and proliferation of autoimmune T cells. One of these two remnant epitopes contains a glycan which is important for its immunoreactivity. In addition to the role of gelatinase B as a regulator in adaptive immune processes, we have also demonstrated that it destroys interferon-beta, a typical innate immunity effector molecule and therapeutic cytokine in multiple sclerosis. Furthermore, glycosylated interferon-beta, expressed in Chinese hamster ovary cells, was more resistant to this proteolysis than recombinant interferon-beta from bacteria. These data not only prove that glycosylation of proteins is mechanistically important in the pathogenesis of autoimmune diseases, but also show that targeting of glycosylated proteinases or the use of glycosylated cytokines seems also critical for the treatment of autoimmune diseases.


Subject(s)
Autoimmunity , Epitopes , Peptide Hydrolases/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/etiology , Glycoproteins/immunology , Glycosylation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...