Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
J Thromb Haemost ; 12(1): 93-102, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24119206

ABSTRACT

BACKGROUND: Thrombus resolution is a complex process that involves thrombosis, leukocyte-mediated thrombolysis, and the final resolution of inflammation. Activated protein C (APC) is an anticoagulant that also possesses immunoregulatory activities. AIM: In this study, we sought to examine the effects of APC administration on thrombus resolution using a mouse model of deep vein thrombosis by ligating the inferior vena cava (IVC). METHODS: The IVCs of C57BL/6 mice were ligated. Beginning on day 4 post IVC ligation, mice were injected intraperitoneally daily with APC, APC plus an heme oxygenase-1 (HO-1) inhibitor Sn-protoporphyrin IX (SnPP), SnPP alone, or vehicle control. At different time points following surgery, the thrombus-containing IVCs were weighed and then analyzed by use of biochemical assays and histology. RESULTS: Venous thrombi reached maximum size on day 4 post ligation. The APC-treated group exhibited a significant reduction in thrombus weights on day 12 but not on day 7 compared with control mice. The enhanced thrombus resolution in APC-treated mice correlated with an increased HO-1 expression and a reduced interleukin-6 production. No significant difference was found in urokinase-type plasminogen activator, plasminogen activator inhibitor-1, or matrix metalloproteinase-2 and -9 between APC-treated and control mice. Coinjection of the HO-1 inhibitor SnPP abolished the ability of APC to enhance thrombus resolution. CONCLUSIONS: Our data show that APC enhances the resolution of existing venous thrombi via a mechanism that is in part dependent on HO-1, suggesting that APC could be used as a potential treatment for patients with deep vein thrombosis to accelerate thrombus resolution.


Subject(s)
Heme Oxygenase-1/biosynthesis , Protein C/pharmacology , Venous Thrombosis/drug therapy , Animals , Base Sequence , DNA Primers , Enzyme Induction , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred C57BL , Protein C/administration & dosage , Protein C/therapeutic use , Real-Time Polymerase Chain Reaction
3.
Mucosal Immunol ; 3(1): 29-39, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19865078

ABSTRACT

Toll-like receptors (TLRs) and proteinase-activated receptors (PARs) function as innate immune biosensors in mucosal epithelial cells (ECs). We previously reported the functional and physical interactions between TLR4 and PAR(2). We have extended these findings herein by showing the cooperation between PAR(2) and TLR2, TLR3, or TLR4 for activation of nuclear factor-kappaB-dependent signaling in mucosal EC lines. In contrast, activation of PAR(2) negatively regulated TLR3-dependent antiviral pathway, blunting the expression of TLR3/interferon regulatory factor-3 (IRF-3)-driven genes, as well as activation of IRF-3 and STAT1. Consistent with these in vitro observations, PAR(2)(-/-) and TLR4(-/-) mice, which were refractory to footpad edema induced by PAR(2) agonist peptide, were protected from mouse-adapted H1N1 influenza A virus-induced lethality when compared to wild-type (WT) mice. These data support and extend our recently described, novel model of PAR(2)-TLR4 "receptor cooperativity" and highlight the complexity of signaling integration between heterologous innate immune biosensors.


Subject(s)
Epithelial Cells/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Receptor, PAR-2/metabolism , Toll-Like Receptors/metabolism , Animals , Cell Line , Edema , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Interferon Regulatory Factor-3/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/pathology , NF-kappa B/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/physiopathology , STAT1 Transcription Factor/metabolism , Signal Transduction/immunology
4.
Br J Cancer ; 92(4): 760-9, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15685234

ABSTRACT

The Testisin gene (PRSS21) encodes a glycosylphosphatidylinositol (GPI)-linked serine protease that exhibits testis tissue-specific expression. Loss of Testisin has been implicated in testicular tumorigenesis, but its role in testis biology and tumorigenesis is not known. Here we have investigated the role of CpG methylation in Testisin gene inactivation and tested the hypothesis that Testisin may act as a tumour suppressor for testicular tumorigenesis. Using sequence analysis of bisulphite-treated genomic DNA, we find a strong relationship between hypermethylation of a 385 bp 5' CpG rich island of the Testisin gene, and silencing of the Testisin gene in a range of human tumour cell lines and in 100% (eight/eight) of testicular germ cell tumours. We show that treatment of Testisin-negative cell lines with demethylating agents and/or a histone deacetylase inhibitor results in reactivation of Testisin gene expression, implicating hypermethylation in Testisin gene silencing. Stable expression of Testisin in the Testisin-negative Tera-2 testicular cancer line suppressed tumorigenicity as revealed by inhibition of both anchorage-dependent cell growth and tumour formation in an SCID mouse model of testicular tumorigenesis. Together, these data show that loss of Testisin is caused, at least in part, by DNA hypermethylation and histone deacetylation, and suggest a tumour suppressor role for Testisin in testicular tumorigenesis.


Subject(s)
CpG Islands , DNA Methylation , DNA, Neoplasm/metabolism , Gene Silencing , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Testicular Neoplasms/metabolism , Adult , Animals , Cell Line, Tumor , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors , Humans , Immunohistochemistry , Male , Membrane Proteins , Mice , Mice, SCID , Orchiectomy , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Testicular Neoplasms/genetics , Testicular Neoplasms/surgery , Transplantation, Heterologous
5.
Lab Invest ; 81(10): 1403-14, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11598153

ABSTRACT

The urokinase plasminogen activator receptor-associated protein/Endo180 (uPARAP/Endo180) is a newly discovered member of the macrophage mannose receptor family that was reported to interact with ligand-bound urokinase plasminogen activator receptor (uPAR), matrix metalloprotease-13 (MMP-13), and collagen V on the cell surface. We have determined the sites of expression of this novel receptor during murine postimplantation development. uPARAP/Endo180 was expressed in all tissues undergoing primary ossification, including the developing bones of the viscerocranium and calvarium that ossify intramembranously, and developing long bones undergoing endochondral ossification. uPARAP/Endo180 mRNA was expressed by both immature osteoblasts and by mature osteocalcin-producing osteoblasts-osteocytes, and was coexpressed with MMP-13. Interestingly, osteoblasts also expressed uPAR. Besides bone-forming tissues, uPARAP/Endo180 expression was detected only in a mesenchymal condensation of the midbrain and in the developing lungs. The data suggest a function of this novel protease receptor in bone development, possibly mediated through its interactions with uPAR, MMP-13, or collagen V.


Subject(s)
Bone and Bones/physiology , Collagenases/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Mitogen/biosynthesis , Animals , Bone and Bones/embryology , Embryonic and Fetal Development , Female , Immunohistochemistry , Matrix Metalloproteinase 13 , Mice , Osteogenesis/physiology , Pregnancy , Receptors, Urokinase Plasminogen Activator
6.
J Biol Chem ; 276(22): 19027-32, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11259424

ABSTRACT

Membrane type 1-matrix metalloproteinase (MT1-MMP)-mediated activation of MMP-2 is thought to be important in the proteolysis of extracellular matrix in pathological events in which monocytes/macrophages are found. Here we report on the induction and regulation of human monocyte MT1-MMP and its role in MMP-2 activation. Activation of monocytes by lipopolysaccharide resulted in the induction of MT1-MMP mRNA and protein that was suppressed by inhibitors of prostaglandin synthesis (indomethacin), adenylyl cyclase (SQ 22536), and protein kinase A (Rp-cAMPs). Suppression of MT1-MMP by indomethacin and SQ 22536 was reversed by prostaglandin E(2) and dibutyryl cyclic AMP, respectively, demonstrating that induction of monocyte MT1-MMP is regulated through a prostaglandin-cAMP pathway. Functional analysis revealed that pro-MMP-2 in the supernatants from human bone marrow stromal fibroblasts, normal male-derived fibroblasts and melanoma cells (A2058) was converted to active MMP-2 when cultured with activated but not control monocytes. Antibodies against MT1-MMP blocked the activation of MMP-2. Tissue inhibitor of metalloproteinase-2 regulation of MMP-2 activation was shown through the addition of varying amounts of recombinant tissue inhibitor of metalloproteinase-2 with pro-MMP-2 to MT1-MMP-expressing monocytes. These findings demonstrate that activated monocytes express functionally active MT1-MMP that may play a significant role in the activation of MMP-2 produced by other cells and as such influence developmental and pathological conditions.


Subject(s)
Adenine/analogs & derivatives , Matrix Metalloproteinase 2/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/physiology , Monocytes/enzymology , Prostaglandins/metabolism , Adenine/pharmacology , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/pharmacology , Blotting, Western , Bucladesine/pharmacology , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/pharmacology , Dinoprostone/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Flow Cytometry , Humans , Indomethacin/pharmacology , Lipopolysaccharides/pharmacology , Matrix Metalloproteinases, Membrane-Associated , Monocytes/metabolism , RNA/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-2/physiology
7.
Cancer Res ; 60(21): 6061-7, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085528

ABSTRACT

Matrix metalloproteinases (MMPs) are overexpressed in a variety of tumor tissues and cell lines, and their expression is highly correlated to tumor invasion and metastasis. To exploit these characteristics in the design of tumor cell-selective cytotoxins, we constructed two mutated anthrax toxin protective antigen (PA) proteins in which the furin protease cleavage site is replaced by sequences selectively cleaved by MMPs. These MMP-targeted PA proteins were activated rapidly and selectively on the surface of MMP-overexpressing tumor cells. The activated PA proteins caused internalization of a recombinant cytotoxin, FP59, consisting of anthrax toxin lethal factor residues 1-254 fused to the ADP-ribosylation domain of Pseudomonas exotoxin A. The toxicity of the mutated PA proteins for MMP-overexpressing cells was blocked by hydroxamate inhibitors of MMPs, including BB94, and by a tissue inhibitor of matrix metalloproteinases (TIMP-2). The mutated PA proteins killed MMP-overexpressing tumor cells while sparing nontumorigenic normal cells when these were grown together in a coculture model, indicating that PA activation occurred on the tumor cell surface and not in the supernatant. This method of achieving cell-type specificity is conceptually distinct from, and potentially synergistic with, the more common strategy of retargeting a protein toxin by fusion to a growth factor, cytokine, or antibody.


Subject(s)
Antigens, Bacterial , Bacterial Toxins/pharmacokinetics , Bacterial Toxins/toxicity , Matrix Metalloproteinases/metabolism , Amino Acid Sequence , Animals , Bacterial Toxins/genetics , Binding Sites , Biotransformation , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , COS Cells/enzymology , Chlorocebus aethiops , Coculture Techniques , Fibrosarcoma/drug therapy , Fibrosarcoma/enzymology , Humans , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases, Membrane-Associated , Melanoma/drug therapy , Melanoma/enzymology , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/metabolism , Mutation , Tumor Cells, Cultured , Vero Cells/enzymology
10.
Biochemistry ; 32(25): 6427-32, 1993 Jun 29.
Article in English | MEDLINE | ID: mdl-8390857

ABSTRACT

The sequence specificities of human 72-kDa fibroblast gelatinase (type IV collagenase), human 92-kDa neutrophil gelatinase (type IV collagenase), and putative metalloproteinase (PUMP or matrilysin) have been examined by measuring the rate of hydrolysis of over 50 synthetic oligopeptides covering the P4 through P4' subsites of the substrate. The peptides investigated in this paper were those employed in our previous study which systematically examined the sequence specificity of human fibroblast and neutrophil collagenases [Netzel-Arnett et al. (1991) J. Biol. Chem. 266, 6747]. The initial rate of hydrolysis of the P1-P1' bond of each peptide has been measured under first-order conditions ([S0] << KM), and kcat/KM values have been calculated from the initial rates. The specificities of these five metalloproteinases are similar, but distinct, with the largest differences occurring at subsites P1, P1', and P3'. The specificities of the two gelatinases are the most similar to each other. They tolerate only small amino acids such as Gly and Ala in subsite P1. In contrast, larger residues such as Met, Pro, Gln, and Glu are also accommodated well by PUMP. All five enzymes prefer hydrophobic, aliphatic residues in subsite P1'. PUMP exhibits a stronger preference for Leu in this subsite than is shown by the other enzymes. The P3' subsite specificities of the gelatinases and collagenases are very similar but different from those of PUMP which particularly prefers Met in this position. The specificity data from this study allow the design of optimized substrates and selective inhibitors for these metalloproteinases.


Subject(s)
Endopeptidases/metabolism , Extracellular Matrix Proteins , Metalloendopeptidases/metabolism , Oligopeptides/metabolism , Aggrecans , Amino Acid Sequence , Chondroitin Sulfate Proteoglycans/metabolism , Collagen/metabolism , Gelatinases , Humans , Kinetics , Lectins, C-Type , Matrix Metalloproteinase 7 , Molecular Sequence Data , Molecular Weight , Proteoglycans/metabolism , Structure-Activity Relationship , Substrate Specificity
12.
Anal Biochem ; 195(1): 86-92, 1991 May 15.
Article in English | MEDLINE | ID: mdl-1888020

ABSTRACT

Four new fluorogenic heptapeptide substrates have been synthesized with sequences that are optimized for five human matrix metalloproteinases (MMP). All four substrates are similar to one recently reported by Stack and Gray (1989, J. Biol. Chem. 264, 4277-4281) and have the fluorescent Trp residue in subsite P'2 and the dinitrophenol (DNP) quenching group on the N-terminus. The quenching of the Trp fluorescence in the intact substrate is relieved on hydrolysis of the P1-P'1 bond, giving rise to a continuously recording fluorescence assay. The residues placed in subsites P3-P'1 and P'3 have been optimized for each MMP, while Arg has been placed in P'4 to enhance solubility. Thus, DNP-Pro-Leu-Ala-Leu-Trp-Ala-Arg has been prepared as a substrate for fibroblast collagenase, DNP-Pro-Leu-Ala-Tyr-Trp-Ala-Arg for neutrophil collagenase, DNP-Pro-Tyr-Ala-Tyr-Trp-Met-Arg for neutrophil collagenase, DNP-Pro-Tyr-Ala-Tyr-Trp-Met-Arg for stromelysin, and DNP-Pro-Leu-Gly-Met-Trp-Ser-Arg for both 72-kDa fibroblast gelatinase and 92-kDa neutrophil gelatinase. These substrates have been characterized with respect to their composition, solubility, optical and fluorescence spectra, and hydrolysis by their target MMP. The hydrolysis rates rival or exceed those of either their natural protein substrates or other synthetic peptides. The solubility of each substrate in assay buffer exceeds the KM value for each reaction, allowing accurate determination of the kinetic parameters. These new substrates should greatly facilitate kinetic studies of the MMP.


Subject(s)
Metalloendopeptidases/chemistry , Amino Acid Sequence , Humans , Hydrolysis , Kinetics , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Spectrometry, Fluorescence
13.
J Biol Chem ; 266(11): 6747-55, 1991 Apr 15.
Article in English | MEDLINE | ID: mdl-1849891

ABSTRACT

The sequence specificities of human fibroblast and neutrophil collagenases have been investigated by measuring the rate of hydrolysis of 60 synthetic oligopeptides covering the P4 through P'5 subsites of the substrate. The choice of peptides was patterned after both known cleavage sites in noncollagenous proteins and potential cleavage sites (those containing Gly-Ile-Ala, Gly-Leu-Ala, or Gly-Ile-Leu sequences) found in types I, II, III, and IV collagens. The initial rate of hydrolysis of the P1-P'1 bond of each peptide has been measured under first-order conditions ([SO] much less than KM), and kcat/KM values have been calculated from the initial rates. The amino acids in subsites P4 through P'4 all influence the hydrolysis rates for both collagenases. However, the effects of substitutions at each site are distinctive and are consistent with the view that human fibroblast and neutrophil collagenases are homologous but nonidentical enzymes. For peptides with unblocked NH2 and COOH termini, occupancy of subsites P3 through P'3 is necessary for rapid hydrolysis. Compared with the alpha 1(I) cleavage sequence, none of the substitutions investigated at subsites P3, P2, and P'4 produces markedly improved substrates. In contrast, many substitutions at subsites P1, P'1, and P'2 improve specificity. The preferences of both collagenases for alanine in subsite P1 and tryptophan or phenylalanine in subsite P'2, is noteworthy. Human neutrophil collagenase accommodates aromatic residues in subsite P'1 much better than human fibroblast collagenase. The subsite preferences observed for human fibroblast collagenase in these studies agree well with the residues found at cleavage sites in noncollagenous substrates. However, the sequence specificities of these collagenases cannot explain the failure of these enzymes to hydrolyze many potentially cleavable but apparently protected sites in intact collagens. This represents additional support for the notion that the local structure of collagen is important in determining the location of collagenase cleavage sites.


Subject(s)
Collagen/metabolism , Microbial Collagenase/metabolism , Neutrophils/enzymology , Amino Acid Sequence , Collagen/chemistry , Fibroblasts/enzymology , Humans , Kinetics , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/isolation & purification , Peptides/metabolism , Substrate Specificity
14.
Biochemistry ; 29(28): 6670-7, 1990 Jul 17.
Article in English | MEDLINE | ID: mdl-2168739

ABSTRACT

The action of human fibroblast collagenase (HFC) on six substrates of markedly different size, sequence, and conformation, including rat type I collagen, rat alpha 1(I) gelatin, beta-casein, and the three synthetic oligopeptides Gly-Pro-Gln-Gly-Ile-Ala-Gly-Gln, Asp-Val-Ala-Gln-Phe-Val-Leu-Thr-Pro-Gly, and Pro-Val-Gln-Pro-Ile-Gly-Pro-Gln, has been examined. The first peptide is a model for the collagenase cleavage site in the alpha 1(I) chain of type I collagen, while the latter two peptides are models for the autolytic activation and degradation sites in pro-HFC, respectively. The goal of these studies was to assess whether HFC hydrolyzes all of these disparate substrates at the same active site. Individual kinetic parameters for the hydrolysis of all six substrates have been determined. Gel zymography experiments using collagen, gelatin, and casein as substrates show that all three activities are associated solely with HFC rather than impurities. Recombinant HFC expressed in Escherichia coli also exhibits caseinase activity, reinforcing the view that this activity is not due to a contaminating protease from fibroblasts. The ratios of these activities agree within experimental error for several independent HFC preparations and do not change when two additional affinity purification steps are employed. The inhibition of the hydrolysis of these substrates by both 1,10-phenanthroline and Boc-Pro-Leu-Gly-NHOH is identical within experimental error. A series of assays carried out in the presence of pairs of these substrates clearly shows that they compete for the same active site.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Fibroblasts/enzymology , Microbial Collagenase/metabolism , Amino Acid Sequence , Animals , Binding Sites , Caseins/metabolism , Collagen/metabolism , Gelatin/metabolism , Humans , Hydrolysis , Kinetics , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Rats , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...