Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 9(1): 2416, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925830

ABSTRACT

Controlled macrophage differentiation and activation in the initiation and resolution of inflammation is crucial for averting progression to chronic inflammatory and autoimmune diseases. Here we show a negative feedback mechanism for proinflammatory IFN-γ activation of macrophages driven by macrophage-associated matrix metalloproteinase 12 (MMP12). Through C-terminal truncation of IFN-γ at 135Glu↓Leu136 the IFN-γ receptor-binding site was efficiently removed thereby reducing JAK-STAT1 signaling and IFN-γ activation of proinflammatory macrophages. In acute peritonitis this signature was absent in Mmp12 -/- mice and recapitulated in Mmp12 +/+ mice treated with a MMP12-specific inhibitor. Similarly, loss-of-MMP12 increases IFN-γ-dependent proinflammatory markers and iNOS+/MHC class II+ macrophage accumulation with worse lymphadenopathy, arthritic synovitis and lupus glomerulonephritis. In active human systemic lupus erythematosus, MMP12 levels were lower and IFN-γ higher compared to treated patients or healthy individuals. Hence, macrophage proteolytic truncation of IFN-γ attenuates classical activation of macrophages as a prelude for resolving inflammation.


Subject(s)
Interferon-gamma/metabolism , Lupus Nephritis/immunology , Macrophage Activation/immunology , Matrix Metalloproteinase 12/metabolism , Animals , Arthritis/immunology , Arthritis/pathology , Biopsy , Cell Line , Collagen/immunology , Disease Models, Animal , Female , Humans , Kidney/pathology , Lupus Nephritis/pathology , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/chemically induced , Peritonitis/immunology , Peritonitis/pathology , Primary Cell Culture , Proteolysis , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/immunology , THP-1 Cells , Thioglycolates/toxicity
2.
Data Brief ; 7: 299-310, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26981551

ABSTRACT

The data described provide a comprehensive resource for the family-wide active site specificity portrayal of the human matrix metalloproteinase family. We used the high-throughput proteomic technique PICS (Proteomic Identification of protease Cleavage Sites) to comprehensively assay 9 different MMPs. We identified more than 4300 peptide cleavage sites, spanning both the prime and non-prime sides of the scissile peptide bond allowing detailed subsite cooperativity analysis. The proteomic cleavage data were expanded by kinetic analysis using a set of 6 quenched-fluorescent peptide substrates designed using these results. These datasets represent one of the largest specificity profiling efforts with subsequent structural follow up for any protease family and put the spotlight on the specificity similarities and differences of the MMP family. A detailed analysis of this data may be found in Eckhard et al. (2015) [1]. The raw mass spectrometry data and the corresponding metadata have been deposited in PRIDE/ProteomeXchange with the accession number PXD002265.

3.
Matrix Biol ; 49: 37-60, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26407638

ABSTRACT

Secreted and membrane tethered matrix metalloproteinases (MMPs) are key homeostatic proteases regulating the extracellular signaling and structural matrix environment of cells and tissues. For drug targeting of proteases, selectivity for individual molecules is highly desired and can be met by high yield active site specificity profiling. Using the high throughput Proteomic Identification of protease Cleavage Sites (PICS) method to simultaneously profile both the prime and non-prime sides of the cleavage sites of nine human MMPs, we identified more than 4300 cleavages from P6 to P6' in biologically diverse human peptide libraries. MMP specificity and kinetic efficiency were mainly guided by aliphatic and aromatic residues in P1' (with a ~32-93% preference for leucine depending on the MMP), and basic and small residues in P2' and P3', respectively. A wide differential preference for the hallmark P3 proline was found between MMPs ranging from 15 to 46%, yet when combined in the same peptide with the universally preferred P1' leucine, an unexpected negative cooperativity emerged. This was not observed in previous studies, probably due to the paucity of approaches that profile both the prime and non-prime sides together, and the masking of subsite cooperativity effects by global heat maps and iceLogos. These caveats make it critical to check for these biologically highly important effects by fixing all 20 amino acids one-by-one in the respective subsites and thorough assessing of the inferred specificity logo changes. Indeed an analysis of bona fide MEROPS physiological substrate cleavage data revealed that of the 37 natural substrates with either a P3-Pro or a P1'-Leu only 5 shared both features, confirming the PICS data. Upon probing with several new quenched-fluorescent peptides, rationally designed on our specificity data, the negative cooperativity was explained by reduced non-prime side flexibility constraining accommodation of the rigidifying P3 proline with leucine locked in S1'. Similar negative cooperativity between P3 proline and the novel preference for asparagine in P1 cements our conclusion that non-prime side flexibility greatly impacts MMP binding affinity and cleavage efficiency. Thus, unexpected sequence cooperativity consequences were revealed by PICS that uniquely encompasses both the non-prime and prime sides flanking the proteomic-pinpointed scissile bond.


Subject(s)
Matrix Metalloproteinases/chemistry , Matrix Metalloproteinases/metabolism , Peptide Library , Proteomics/methods , Amino Acid Sequence , Catalytic Domain , Chromatography, Liquid , Humans , Models, Molecular , Molecular Docking Simulation , Substrate Specificity , Tandem Mass Spectrometry
4.
Transfusion ; 55 Suppl 2: S13-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26174892

ABSTRACT

BACKGROUND: Increased reporting of intravenous immunoglobulin (IVIG)-related hemolytic reactions (HRs) triggered an investigation by the German and Swiss health authorities to identify potential risk factors. STUDY DESIGN AND METHODS: From the EudraVigilance database HRs reported between 2008 and 2013 were retrieved for seven IVIG preparations. HRs were classified as mild to moderate (hemoglobin [Hb] decline < 2 g/dL)] or severe (Hb decline > 2 g/dL) and separately analyzed for IVIG doses of less than 2 g/kg body weight and 2 g/kg body weight or more. It was assessed whether HR reporting rates correlate with the isoagglutinin content of the different preparations. RESULTS: Of 569 HR cases retrieved, 103 cases were excluded due to insufficient data, leaving 466 for analysis. Ninety-three cases were classified as mild to moderate and 373 as severe. Approximately 80% of the severe HRs concerned patients with blood group A and only three patients with blood group O. Testing of isoagglutinin titers revealed substantial differences between the seven preparations. IVIG products with high anti-A/anti-B titers (≥32) had elevated HR reporting rates, particularly when cumulative doses at least 2 g/kg were administered. CONCLUSION: The isoagglutinin content of IVIGs correlates with the risk for HRs. Exclusion of high-titer donations and manufacturing steps that deplete isoagglutinins should be considered for risk mitigation. In patients with blood groups A or AB receiving doses of at least 2 g/kg, the use of IVIG batches with low isoagglutinin titers should be considered to prevent HRs.


Subject(s)
ABO Blood-Group System/blood , Databases, Factual , Hemagglutinins/adverse effects , Immunoglobulins, Intravenous/adverse effects , Isoantibodies/adverse effects , Female , Hemagglutinins/administration & dosage , Hemagglutinins/chemistry , Hemoglobins/metabolism , Humans , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/chemistry , Isoantibodies/administration & dosage , Isoantibodies/chemistry , Male , Risk Factors
5.
Cell Rep ; 9(2): 618-32, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25310974

ABSTRACT

Resolution of inflammation reduces pathological tissue destruction and restores tissue homeostasis. Here, we used a proteomic protease substrate discovery approach, terminal amine isotopic labeling of substrates (TAILS), to analyze the role of the macrophage-specific matrix metalloproteinase-12 (MMP12) in inflammation. In murine peritonitis, MMP12 inactivates antithrombin and activates prothrombin, prolonging the activated partial thromboplastin time. Furthermore, MMP12 inactivates complement C3 to reduce complement activation and inactivates the chemoattractant anaphylatoxins C3a and C5a, whereas iC3b and C3b opsonin cleavage increases phagocytosis. Loss of these anti-inflammatory activities in collagen-induced arthritis in Mmp12(-/-) mice leads to unresolved synovitis and extensive articular inflammation. Deep articular cartilage loss is associated with massive neutrophil infiltration and abnormal DNA neutrophil extracellular traps (NETs). The NETs are rich in fibrin and extracellular actin, which TAILS identified as MMP12 substrates. Thus, macrophage MMP12 in arthritis has multiple protective roles in countering neutrophil infiltration, clearing NETs, and dampening inflammatory pathways to prepare for the resolution of inflammation.


Subject(s)
Arthritis, Experimental/metabolism , Macrophages/metabolism , Matrix Metalloproteinase 12/metabolism , Neutrophil Infiltration , Neutrophils/immunology , Actins/metabolism , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cartilage/pathology , Cell Line , Complement Activation , Complement C3/immunology , Extracellular Traps/metabolism , Fibrin/metabolism , Male , Matrix Metalloproteinase 12/genetics , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Peritonitis/immunology , Peritonitis/metabolism , Prothrombin/metabolism
6.
J Neuroinflammation ; 11: 169, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25274277

ABSTRACT

BACKGROUND: Bacterial meningitis (BM) is characterized by an intense host inflammatory reaction, which contributes to the development of brain damage and neuronal sequelae. Activation of the kynurenine (KYN) pathway (KP) has been reported in various neurological diseases as a consequence of inflammation. Previously, the KP was shown to be activated in animal models of BM, and the association of the SNP AADAT + 401C/T (kynurenine aminotransferase II - KAT II) with the host immune response to BM has been described. The aim of this study was to investigate the involvement of the KP during BM in humans by assessing the concentrations of KYN metabolites in the cerebrospinal fluid (CSF) of BM patients and their relationship with the inflammatory response compared to aseptic meningitis (AM) and non-meningitis (NM) groups. METHODS: The concentrations of tryptophan (TRP), KYN, kynurenic acid (KYNA) and anthranilic acid (AA) were assessed by HPLC from CSF samples of patients hospitalized in the Giselda Trigueiro Hospital in Natal (Rio Grande do Norte, Brazil). The KYN/TRP ratio was used as an index of indoleamine 2,3-dioxygenase (IDO) activity, and cytokines were measured using a multiplex cytokine assay. The KYNA level was also analyzed in relation to AADAT + 401C/T genotypes. RESULTS: In CSF from patients with BM, elevated levels of KYN, KYNA, AA, IDO activity and cytokines were observed. The cytokines INF-γ and IL-1Ra showed a positive correlation with IDO activity, and TNF-α and IL-10 were positively correlated with KYN and KYNA, respectively. Furthermore, the highest levels of KYNA were associated with the AADAT + 401 C/T variant allele. CONCLUSION: This study suggests a downward modulatory effect of the KP on CSF inflammation during BM.


Subject(s)
Cytokines/cerebrospinal fluid , Inflammation/cerebrospinal fluid , Kynurenine/cerebrospinal fluid , Meningitis, Bacterial/cerebrospinal fluid , Adolescent , Adult , Chromatography, High Pressure Liquid , Female , Humans , Male , Young Adult
7.
Nat Med ; 20(5): 493-502, 2014 May.
Article in English | MEDLINE | ID: mdl-24784232

ABSTRACT

Interferon-α (IFN-α) is essential for antiviral immunity, but in the absence of matrix metalloproteinase-12 (MMP-12) or IκBα (encoded by NFKBIA) we show that IFN-α is retained in the cytosol of virus-infected cells and is not secreted. Our findings suggest that activated IκBα mediates the export of IFN-α from virus-infected cells and that the inability of cells in Mmp12(-/-) but not wild-type mice to express IκBα and thus export IFN-α makes coxsackievirus type B3 infection lethal and renders respiratory syncytial virus more pathogenic. We show here that after macrophage secretion, MMP-12 is transported into virus-infected cells. In HeLa cells MMP-12 is also translocated to the nucleus, where it binds to the NFKBIA promoter, driving transcription. We also identified dual-regulated substrates that are repressed both by MMP-12 binding to the substrate's gene exons and by MMP-12-mediated cleavage of the substrate protein itself. Whereas intracellular MMP-12 mediates NFKBIA transcription, leading to IFN-α secretion and host protection, extracellular MMP-12 cleaves off the IFN-α receptor 2 binding site of systemic IFN-α, preventing an unchecked immune response. Consistent with an unexpected role for MMP-12 in clearing systemic IFN-α, treatment of coxsackievirus type B3-infected wild-type mice with a membrane-impermeable MMP-12 inhibitor elevates systemic IFN-α levels and reduces viral replication in pancreas while sparing intracellular MMP-12. These findings suggest that inhibiting extracellular MMP-12 could be a new avenue for the development of antiviral treatments.


Subject(s)
Cell Nucleus/genetics , Immunity/genetics , Interferon-alpha/genetics , Matrix Metalloproteinase 12/genetics , Animals , Binding Sites , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cytosol/metabolism , Cytosol/virology , HeLa Cells , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Interferon-alpha/immunology , Interferon-alpha/metabolism , Matrix Metalloproteinase 12/metabolism , Mice , Mice, Knockout , NF-KappaB Inhibitor alpha , Pancreas/immunology , Pancreas/virology , Rous sarcoma virus/genetics , Rous sarcoma virus/pathogenicity , Virus Replication/drug effects
8.
BMC Infect Dis ; 13: 393, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23977941

ABSTRACT

BACKGROUND: Bacterial meningitis caused by Streptococcus pneumoniae leads to death in up to 30% of patients and leaves up to half of the survivors with neurological sequelae. The inflammatory host reaction initiates the induction of the kynurenine pathway and contributes to hippocampal apoptosis, a form of brain damage that is associated with learning and memory deficits in experimental paradigms. Vitamin B6 is an enzymatic cofactor in the kynurenine pathway and may thus limit the accumulation of neurotoxic metabolites and preserve the cellular energy status. The aim of this study in a pneumococcal meningitis model was to investigate the effect of vitamin B6 on hippocampal apoptosis by histomorphology, by transcriptomics and by measurement of cellular nicotine amide adenine dinucleotide content. METHODS AND RESULTS: Eleven day old Wistar rats were infected with 1x10(6) cfu/ml of S. pneumoniae and randomized for treatment with vitamin B6 or saline as controls. Vitamin B6 led to a significant (p > 0.02) reduction of hippocampal apoptosis. According to functional annotation based clustering, vitamin B6 led to down-regulation of genes involved in processes of inflammatory response, while genes encoding for processes related to circadian rhythm, neuronal signaling and apoptotic cell death were mostly up-regulated. CONCLUSIONS: Our results provide evidence that attenuation of apoptosis by vitamin B6 is multi-factorial including down-modulation of inflammation, up-regulation of the neuroprotective brain-derived neurotrophic factor and prevention of the exhaustion of cellular energy stores. The neuroprotective effect identifies vitamin B6 as a potential target for the development of strategies to attenuate brain injury in bacterial meningitis.


Subject(s)
Apoptosis/drug effects , Hippocampus/drug effects , Meningitis, Pneumococcal/drug therapy , Vitamin B 6/administration & dosage , Animals , Female , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Meningitis, Pneumococcal/genetics , Meningitis, Pneumococcal/metabolism , Meningitis, Pneumococcal/physiopathology , Rats , Rats, Wistar , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/isolation & purification , Streptococcus pneumoniae/physiology
9.
Cell Mol Life Sci ; 70(2): 309-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22940918

ABSTRACT

The in vivo roles of meprin metalloproteases in pathophysiological conditions remain elusive. Substrates define protease roles. Therefore, to identify natural substrates for human meprin α and ß we employed TAILS (terminal amine isotopic labeling of substrates), a proteomics approach that enriches for N-terminal peptides of proteins and cleavage fragments. Of the 151 new extracellular substrates we identified, it was notable that ADAM10 (a disintegrin and metalloprotease domain-containing protein 10)-the constitutive α-secretase-is activated by meprin ß through cleavage of the propeptide. To validate this cleavage event, we expressed recombinant proADAM10 and after preincubation with meprin ß, this resulted in significantly elevated ADAM10 activity. Cellular expression in murine primary fibroblasts confirmed activation. Other novel substrates including extracellular matrix proteins, growth factors and inhibitors were validated by western analyses and enzyme activity assays with Edman sequencing confirming the exact cleavage sites identified by TAILS. Cleavages in vivo were confirmed by comparing wild-type and meprin(-/-) mice. Our finding of cystatin C, elafin and fetuin-A as substrates and natural inhibitors for meprins reveal new mechanisms in the regulation of protease activity important for understanding pathophysiological processes.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Extracellular Matrix Proteins/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , ADAM10 Protein , Amino Acid Sequence , Animals , Caco-2 Cells , Cell Line , Cystatin C/metabolism , Cytokines/metabolism , Elafin/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , alpha-2-HS-Glycoprotein/metabolism
10.
J Biol Chem ; 287(16): 13382-95, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22367194

ABSTRACT

The neutrophil-specific protease membrane-type 6 matrix metalloproteinase (MT6-MMP)/MMP-25/leukolysin is implicated in multiple sclerosis and cancer yet remains poorly characterized. To characterize the biological roles of MT6-MMP, it is critical to identify its substrates for which only seven are currently known. Here, we biochemically characterized MT6-MMP, profiled its tissue inhibitor of metalloproteinase inhibitory spectrum, performed degradomics analyses, and screened 26 chemokines for cleavage using matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry. MT6-MMP processes seven each of the CXC and CC chemokine subfamilies. Notably, cleavage of the neutrophil chemoattractant CXCL5 activates the chemokine, thereby increasing its agonist activity, indicating a feed-forward mechanism for neutrophil recruitment. Likewise, cleavage also activated CCL15 and CCL23 to increase monocyte recruitment. Utilizing the proteomics approach proteomic identification of cleavage site specificity (PICS), we identified 286 peptidic cleavage sites spanning from P6 to P6' from which an unusual glutamate preference in P1 was identified. The degradomics screen terminal amine isotopic labeling of substrates (TAILS), which enriches for neo-N-terminal peptides of cleaved substrates, was used to identify 58 new native substrates in fibroblast secretomes after incubation with MT6-MMP. Vimentin, cystatin C, galectin-1, IGFBP-7, and secreted protein, acidic and rich in cysteine (SPARC) were among those substrates we biochemically confirmed. An extracellular "moonlighting" form of vimentin is a chemoattractant for THP-1 cells, but MT6-MMP cleavage abolished monocyte recruitment. Unexpectedly, the MT6-MMP-cleaved vimentin potently stimulated phagocytosis, which was not a property of the full-length protein. Hence, MT6-MMP regulates neutrophil and monocyte chemotaxis and by generating "eat-me" signals upon vimentin cleavage potentially increases phagocytic removal of neutrophils to resolve inflammation.


Subject(s)
Cell Movement/immunology , Chemokines/metabolism , Macrophages/immunology , Matrix Metalloproteinases, Membrane-Associated , Phagocytosis/immunology , Vimentin/metabolism , Amino Acid Sequence , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , Immunity, Innate/immunology , K562 Cells , Macrophages/cytology , Matrix Metalloproteinases, Membrane-Associated/genetics , Matrix Metalloproteinases, Membrane-Associated/immunology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Molecular Sequence Data , Neutrophils/cytology , Neutrophils/immunology , Proteomics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Substrate Specificity/immunology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Vimentin/genetics
11.
J Neuropathol Exp Neurol ; 69(11): 1096-104, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20940631

ABSTRACT

Pneumococcal meningitis causes neurological sequelae, including learning and memory deficits in up to half of the survivors. In both humans and in animal models of the disease, there is apoptotic cell death in the hippocampus, a brain region involved in learning and memory function. We previously demonstrated that in an infant rat model of pneumococcal meningitis, there is activation of the kynurenine (KYN) pathway in the hippocampus, and that there was a positive correlation between the concentration of 3-hydroxykynurenine and the extent of hippocampal apoptosis. To clarify the role of the KYN pathway in the pathogenesis of hippocampal apoptosis in pneumococcal meningitis, we specifically inhibited 2 key enzymes of the KYN pathway and assessed hippocampal apoptosis, KYN pathway metabolites, and nicotinamide adenine dinucleotide (NAD) concentrations by high-performance liquid chromatography. Pharmacological inhibition of kynurenine 3-hydroxylase and kynureninase led to decreased cellular NAD levels and increased apoptosis in the hippocampus. The cerebrospinal fluid levels of tumor necrosis factor and interleukin-1α and -ß were not affected. Our data suggest that activation of the KYN pathway in pneumococcal meningitis is neuroprotective by compensating for an increased NAD demand caused by infection and inflammation;this mechanism may prevent energy failure and apoptosis in the hippocampus.


Subject(s)
Apoptosis/physiology , Energy Metabolism/physiology , Kynurenine/metabolism , Meningitis, Pneumococcal/pathology , NAD/metabolism , Signal Transduction/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Chromatography, High Pressure Liquid/methods , Cytokines/cerebrospinal fluid , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Energy Metabolism/drug effects , Hippocampus/metabolism , Meningitis, Pneumococcal/metabolism , Meningitis, Pneumococcal/physiopathology , Myelin Basic Protein/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thiazoles/pharmacology
12.
Cancer Res ; 70(19): 7562-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20729277

ABSTRACT

Matrix metalloproteinases (MMP), strongly associated pathogenic markers of cancer, have undergone extensive drug development programs. Marimastat, a noncovalent MMP inhibitor, was conjugated with FITC to label cellular metalloproteinase cancer targets in MDA-MB-231 cells in vitro. Punctate localization of active transmembrane MMP14 was observed. For molecular-targeted positron emission tomography imaging of syngeneic 67NR murine mammary carcinoma in vivo, marimastat was (18)F-labeled using a shelf-stable arylboronic ester conjugate as a captor for aqueous [(18)F]fluoride in a novel, rapid one-step reaction at ambient temperature. [(18)F]Marimastat-aryltrifluoroborate localized to the tumors, with labeling being blocked in control animals first loaded with >10-fold excess unlabeled marimastat. The labeled drug cleared primarily via the hepatobiliary and gastrointestinal tract, with multiple animals imaged in independent experiments, confirming the ease of this new labeling strategy.


Subject(s)
Breast Neoplasms/diagnostic imaging , Enzyme Inhibitors , Fluorine Radioisotopes , Hydroxamic Acids , Mammary Neoplasms, Experimental/diagnostic imaging , Matrix Metalloproteinase Inhibitors , Radiopharmaceuticals , Animals , Borates/chemistry , Breast Neoplasms/enzymology , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Female , Fluorine Radioisotopes/chemistry , Humans , Hydroxamic Acids/chemistry , Isoenzymes , Isotope Labeling/methods , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Inbred BALB C , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemical synthesis
13.
Blood ; 112(8): 3455-64, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18660381

ABSTRACT

Through the activity of macrophage-specific matrix metalloproteinase-12 (MMP-12), we found that macrophages dampen the lipopolysaccharide (LPS)-induced influx of polymorphonuclear leukocytes (PMNs)-thus providing a new mechanism for the termination of PMN recruitment in acute inflammation. MMP-12 specifically cleaves human ELR(+) CXC chemokines (CXCL1, -2, -3, -5, and -8) at E-LR, the critical receptor-binding motif or, for CXCL6, carboxyl-terminal to it. Murine (m) MMP-12 also cleaves mCXCL1, -2, and -3 at E-LR. MMP-12-cleaved mCXCL2 (macrophage-inflammatory protein-2 [MIP-2]) and mCXCL3 (dendritic cell inflammatory protein-1 [DCIP-1]) lost chemotactic activity. Furthermore, MMP-12 processed and inactivated monocyte chemotactic proteins CCL2, -7, -8, and -13 at position 4-5 generating CCR antagonists. Indeed, PMNs and macrophages in bronchoalveolar lavage fluid were significantly increased 72 hours after intranasal instillation of LPS in Mmp12(-/-) mice compared with wild type. Specificity occurred at 2 levels. Macrophage MMP-1 and MMP-9 did not cleave in the ELR motif. Second, unlike human ELR(+)CXC chemokines, mCXCL5 (LPS-induced CXC chemokine [LIX]) was not inactivated. Rather, mMMP-12 cleavage at Ser4-Val5 activated the chemokine, promoting enhanced PMN early infiltration in wild-type mice compared with Mmp12(-/-) mice 8 hours after LPS challenge in air pouches. We propose that the macrophage, specifically through MMP-12, assists in orchestrating the regulation of acute inflammatory responses by precise proteolysis of ELR(+)CXC and CC chemokines.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , Chemokine CCL8/metabolism , Chemokines, CXC/metabolism , Gene Expression Regulation , Macrophages/metabolism , Matrix Metalloproteinase 12/physiology , Monocyte Chemoattractant Proteins/metabolism , Neutrophils/metabolism , Amino Acid Motifs , Animals , Cell Movement , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL7/antagonists & inhibitors , Chemokine CCL8/antagonists & inhibitors , Female , Humans , Male , Matrix Metalloproteinase 12/metabolism , Mice , Mice, Transgenic , Monocyte Chemoattractant Proteins/antagonists & inhibitors , Neutrophils/cytology
14.
Neurobiol Dis ; 28(2): 175-83, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17706429

ABSTRACT

Inflammation of the subarachnoid and ventricular space contributes to the development of brain damage i.e. cortical necrosis and hippocampal apoptosis in pneumococcal meningitis (PM). Galectin-3 and -9 are known pro-inflammatory mediators and regulators of apoptosis. Here, the gene and protein expression profile for both galectins was assessed in the disease progression of PM. The mRNA of Lgals3 and Lgals9 increased continuously in the cortex and in the hippocampus from 22 h to 44 h after infection. At 44 h after infection, mRNA levels of Lgals9 in the hippocampus were 7-fold and those of Lgals3 were 30-fold higher than in uninfected controls (P<0.01). Galectin-9 protein did not change, but galectin-3 significantly increased in cortex and hippocampus with the duration of PM. Galectin-3 was localized to polymorphonuclear neutrophils, microglia, monocytes and macrophages, suggesting an involvement of galectin-3 in the neuroinflammatory processes leading to brain damage in PM.


Subject(s)
Brain/metabolism , Chemotaxis, Leukocyte/immunology , Encephalitis/metabolism , Galectin 3/metabolism , Galectins/metabolism , Meningitis, Pneumococcal/metabolism , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/immunology , Antigens, Differentiation, Myelomonocytic/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Brain/immunology , Brain/microbiology , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/microbiology , Chemotaxis, Leukocyte/genetics , Disease Models, Animal , Encephalitis/genetics , Encephalitis/microbiology , Galectin 3/genetics , Galectin 3/immunology , Galectins/genetics , Galectins/immunology , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/microbiology , Macrophages/immunology , Macrophages/metabolism , Meningitis, Pneumococcal/genetics , Meningitis, Pneumococcal/physiopathology , Microglia/immunology , Microglia/metabolism , Monocytes/immunology , Monocytes/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Neutrophils/immunology , Neutrophils/metabolism , RNA, Messenger/metabolism , Rats , Up-Regulation/genetics , Up-Regulation/immunology
15.
Neurobiol Dis ; 24(2): 395-402, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16956766

ABSTRACT

Pneumococcal meningitis (PM) is characterized by an intense inflammatory host reaction that contributes to the development of cortical necrosis and hippocampal apoptosis. Inflammatory conditions in the brain are known to induce tryptophan degradation along the kynurenine pathway, resulting in accumulation of neurotoxic metabolites. In the present study, we investigated the contribution of the kynurenine pathway to brain injury in experimental PM by measuring the concentration of its metabolites and the enzymatic activities and mRNA levels of its major enzymes in the vulnerable brain regions. In the late phase of acute PM, we found a significant transcriptional upregulation of kynurenine-3-hydroxylase and an accumulation of the neurotoxic metabolites 3-hydroxykynurenine (3-HKYN) and 3-hydroxyanthranilic acid in cortex and hippocampus. The positive correlation between the concentration of 3-HKYN and the extent of hippocampal apoptosis adds support to the concept that 3-HKYN contributes to brain injury in PM.


Subject(s)
Brain/metabolism , Encephalitis/metabolism , Kynurenine/metabolism , Meningitis, Pneumococcal/metabolism , Nerve Degeneration/metabolism , Neurotoxins/biosynthesis , 3-Hydroxyanthranilic Acid/metabolism , Animals , Apoptosis/physiology , Brain/microbiology , Brain/physiopathology , Disease Models, Animal , Encephalitis/microbiology , Encephalitis/physiopathology , Enzyme Activation/physiology , Hippocampus/metabolism , Hippocampus/microbiology , Hippocampus/physiopathology , Kynurenine/analogs & derivatives , Kynurenine 3-Monooxygenase/metabolism , Meningitis, Pneumococcal/physiopathology , Nerve Degeneration/microbiology , Nerve Degeneration/physiopathology , Rats , Rats, Wistar , Up-Regulation/physiology
16.
Infect Immun ; 74(7): 3890-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16790761

ABSTRACT

Bacterial meningitis is characterized by an inflammatory reaction to the invading pathogens that can ultimately lead to sensorineural hearing loss, permanent brain injury, or death. The matrix metalloproteinases (MMPs) and tumor necrosis factor alpha-converting enzyme (TACE) are key mediators that promote inflammation, blood-brain barrier disruption, and brain injury in bacterial meningitis. Doxycycline is a clinically used antibiotic with anti-inflammatory effects that lead to reduced cytokine release and the inhibition of MMPs. Here, doxycycline inhibited TACE with a 50% inhibitory dose of 74 microM in vitro and reduced the amount of tumor necrosis factor alpha released into the cerebrospinal fluid by 90% in vivo. In an infant rat model of pneumococcal meningitis, a single dose of doxycycline (30 mg/kg) given as adjuvant therapy in addition to ceftriaxone 18 h after infection significantly reduced the mortality, the blood-brain barrier disruption, and the extent of cortical brain injury. Adjuvant doxycycline (30 mg/kg given subcutaneously once daily for 4 days) also attenuated hearing loss, as assessed by auditory brainstem response audiometry, and neuronal death in the cochlear spiral ganglion at 3 weeks after infection. Thus, doxycycline, probably as a result of its anti-inflammatory properties, had broad beneficial effects in the brain and the cochlea and improved survival in this model of pneumococcal meningitis in infant rats.


Subject(s)
Anti-Bacterial Agents/pharmacology , Brain/microbiology , Brain/pathology , Cochlea/microbiology , Cochlea/pathology , Doxycycline/pharmacology , Meningitis, Pneumococcal/drug therapy , Meningitis, Pneumococcal/mortality , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ceftriaxone/antagonists & inhibitors , Ceftriaxone/pharmacology , Doxycycline/pharmacokinetics , Female , Injections, Subcutaneous , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...