Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 285(10): 7384-93, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20048148

ABSTRACT

Pro-lysyl oxidase is secreted as a 50-kDa proenzyme and is then cleaved to a 30-kDa mature enzyme (lysyl oxidase (LOX)) and an 18-kDa propeptide (lysyl oxidase propeptide (LOX-PP)). The presence of LOX-PP in the cell layers of phenotypically normal osteoblast cultures led us to investigate the effects of LOX-PP on osteoblast differentiation. Data indicate that LOX-PP inhibits terminal mineralization in primary calvaria osteoblast cultures when added at early stages of differentiation, with no effects seen when present at later stages. LOX-PP was found to inhibit serum- and FGF-2-stimulated DNA synthesis and FGF-2-stimulated cell growth. Enzyme-linked immunosorbent assay and Western blot analyses show that LOX-PP inhibits FGF-2-induced ERK1/2 phosphorylation, signaling events that mediate the FGF-2-induced proliferative response. LOX-PP inhibits FGF-2-stimulated phosphorylation of FRS2alpha and FGF-2-stimulated DNA synthesis, even after inhibition of sulfation of heparan sulfate proteoglycans. These data point to a LOX-PP target at or near the level of fibroblast growth factor receptor binding or activation. Ligand binding assays on osteoblast cell layers with (125)I-FGF-2 demonstrate a concentration-dependent inhibition of FGF-2 binding to osteoblasts by LOX-PP. In vitro binding assays with recombinant fibroblast growth factor receptor protein revealed that LOX-PP inhibits FGF-2 binding in an uncompetitive manner. We propose a working model for the respective roles of LOX enzyme and LOX-PP in osteoblast phenotype development in which LOX-PP may act to inhibit the proliferative response possibly to allow cells to exit from the cell cycle and progress to the next stages of differentiation.


Subject(s)
Cell Proliferation , Fibroblast Growth Factor 2/metabolism , Osteoblasts/physiology , Protein Precursors/metabolism , Protein-Lysine 6-Oxidase/metabolism , Signal Transduction/physiology , 3T3 Cells , Animals , Calcification, Physiologic/drug effects , Calcification, Physiologic/physiology , Cattle , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , DNA/biosynthesis , Fibroblast Growth Factor 2/genetics , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/physiology , Protein Precursors/genetics , Protein Precursors/pharmacology , Protein-Lysine 6-Oxidase/genetics , Radioligand Assay , Rats , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Skull/cytology
2.
Biochem Biophys Res Commun ; 366(1): 156-61, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18060869

ABSTRACT

Lysyl oxidase is required for the normal biosynthesis and maturation of collagen and elastin. It is expressed by vascular smooth muscle cells, and its increased expression has been previously found in atherosclerosis and in models of balloon angioplasty. The lysyl oxidase propeptide (LOX-PP) has more recently been found to have biological activity as a tumor suppressor, and it inhibits Erk1/2 Map kinase activation. We reasoned that LOX-PP may have functions in normal non-transformed cells. We, therefore, investigated its effects on smooth muscle cells, focusing on important biological processes mediated by Erk1/2-dependent signaling pathways including proliferation and matrix metalloproteinase-9 (MMP-9) expression. In addition, we investigated whether evidence for accumulation of LOX-PP could be found in vivo in a femoral artery injury model. Recombinant LOX-PP was expressed and purified, and was found to inhibit primary rat aorta smooth muscle cell proliferation and DNA synthesis by more than 50%. TNF-alpha-stimulated MMP-9 expression and Erk1/2 activation were both significantly inhibited by LOX-PP. Immunohistochemistry studies carried out with affinity purified anti-LOX-PP antibody showed that LOX-PP epitopes were expressed at elevated levels in vascular lesions of injured arteries. These novel data suggest that LOX-PP may provide a feedback control mechanism that serves to inhibit properties associated with the development of vascular pathology.


Subject(s)
Cell Proliferation/drug effects , Myocytes, Smooth Muscle/physiology , Peptides/administration & dosage , Protein-Lysine 6-Oxidase/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Rats , Signal Transduction/drug effects
3.
J Biol Chem ; 282(21): 15416-29, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17428796

ABSTRACT

Prostaglandin E(2) blocks transforming growth factor TGF beta1-induced CCN2/CTGF expression in lung and kidney fibroblasts. PGE(2) levels are high in gingival tissues yet CCN2/CTGF expression is elevated in fibrotic gingival overgrowth. Gingival fibroblast expression of CCN2/CTGF in the presence of PGE(2) led us to compare the regulation of CCN2/CTGF expression in fibroblasts cultured from different tissues. Data demonstrate that the TGFbeta1-induced expression of CCN2/CTGF in human lung and renal mesangial cells is inhibited by 10 nm PGE(2), whereas human gingival fibroblasts are resistant. Ten nm PGE(2) increases cAMP accumulation in lung but not gingival fibroblasts, which require 1 mum PGE(2) to elevate cAMP. Micromolar PGE(2) only slightly reduces the TGFbeta1-stimulated CCN2/CTGF levels in gingival cells. EP2 prostaglandin receptor activation with butaprost blocks the TGFbeta1-stimulated expression of CCN2/CTGF expression in lung, but not gingival, fibroblasts. In lung fibroblasts, inhibition of the TGFbeta1-stimulated CCN2/CTGF by PGE(2), butaprost, or forskolin is due to p38, ERK, and JNK MAP kinase inhibition that is cAMP-dependent. Inhibition of any two MAPKs completely blocks CCN2/CTGF expression stimulated by TGFbeta1. These data mimic the inhibitory effects of 10 nm PGE(2) and forskolin that were dependent on PKA activity. In gingival fibroblasts, the sole MAPK mediating the TGFbeta1-stimulated CCN2/CTGF expression is JNK. Whereas forskolin reduces TGFbeta1-stimulated expression of CCN2/CTGF by 35% and JNK activation in gingival fibroblasts, micromolar PGE(2)-stimulated JNK in gingival fibroblasts and opposes the inhibitory effects of cAMP on CCN2/CTGF expression. Stimulation of the EP3 receptor with sulprostone results in a robust increase in JNK activation in these cells. Taken together, data identify two mechanisms by which TGFbeta1-stimulated CCN2/CTGF levels in human gingival fibroblasts resist down-regulation by PGE(2): (i) cAMP cross-talk with MAPK pathways is limited in gingival fibroblasts; (ii) PGE(2) activation of the EP3 prostanoid receptor stimulates the activation of JNK.


Subject(s)
Cyclic AMP/metabolism , Dinoprostone/pharmacology , Fibroblasts/metabolism , Gingiva/metabolism , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Receptors, Prostaglandin E/metabolism , Transforming Growth Factor beta1/pharmacology , Adult , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Cells, Cultured , Colforsin/pharmacology , Connective Tissue Growth Factor , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fibroblasts/cytology , Fibrosis , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gingiva/cytology , Humans , Lung/cytology , Lung/metabolism , MAP Kinase Signaling System/physiology , Mesangial Cells/cytology , Mesangial Cells/metabolism , Organ Specificity/physiology , Receptors, Prostaglandin E, EP3 Subtype , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Am J Physiol Cell Physiol ; 292(6): C2095-102, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17287363

ABSTRACT

Lysyl oxidase plays a critical role in the formation of the extracellular matrix, and its activity is required for the normal maturation and cross-linking of collagen and elastin. An 18-kDa lysyl oxidase propeptide (LOPP) is generated from 50-kDa prolysyl oxidase by extracellular proteolytic cleavage during the biosynthesis of active 30-kDa lysyl oxidase enzyme. The fate and the functions of the LOPP are largely unknown, although intact LOPP was previously observed in osteoblast cultures. We investigated the spatial localization of molecular forms of lysyl oxidase, including LOPP in proliferating and differentiating osteoblasts, by using confocal immunofluorescence microscopy and Western blots of cytoplasmic and nuclear extracts. In the present study, a stage-dependent intracellular distribution of LOPP in the osteoblastic cell was observed. In proliferating osteoblasts, LOPP epitopes were principally associated with the Golgi and endoplasmic reticulum, and mature lysyl oxidase epitopes were found principally in the nucleus and perinuclear region. In differentiating cells, LOPP and mature lysyl oxidase immunostaining showed clear colocalization with the microtubule network. The subcellular distribution of LOPP and its temporal and physical association with microtubules were confirmed by Western blot and far Western blot studies. We also report that N-glycosylated and nonglycosylated LOPP are present in MC3T3-E1 cell cultures. We conclude that LOPP has a stage-dependent intracellular distribution in osteoblastic cells. Future studies are needed to investigate whether the LOPP associations with microtubules or the osteoblast nucleus have functional effects for osteoblast differentiation and bone formation.


Subject(s)
Osteoblasts/cytology , Osteoblasts/metabolism , Protein-Lysine 6-Oxidase/metabolism , 3T3 Cells , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression Regulation, Enzymologic , Mice , Protein Transport , Protein-Lysine 6-Oxidase/genetics , Tubulin
5.
Cancer Res ; 67(3): 1105-12, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17283144

ABSTRACT

Expression of the lysyl oxidase gene (LOX) was found to inhibit the transforming activity of the ras oncogene in NIH 3T3 fibroblasts and was hence named the ras recision gene (rrg). Lysyl oxidase (LOX) is synthesized and secreted as a 50-kDa inactive proenzyme (Pro-LOX), which is processed by proteolytic cleavage to a functional 32-kDa enzyme and an 18-kDa propeptide (LOX-PP). Recently, the ras recision activity of the LOX gene in NIH 3T3 cells was mapped to its propeptide region. Here, we show for the first time that LOX-PP inhibits transformation of breast cancer cells driven by Her-2/neu, an upstream activator of Ras. LOX-PP expression in Her-2/neu-driven breast cancer cells in culture suppressed Akt, extracellular signal-regulated kinase, and nuclear factor-kappaB activation. Her-2/neu-induced epithelial to mesenchymal transition was reverted by LOX-PP, as judged by reduced levels of Snail and vimentin; up-regulation of E-cadherin, gamma-catenin, and estrogen receptor alpha; and decreased ability to migrate or to form branching colonies in Matrigel. Furthermore, LOX-PP inhibited Her-2/neu tumor formation in a nude mouse xenograft model. Thus, LOX-PP inhibits signaling cascades induced by Her-2/neu that promote a more invasive phenotype and may provide a novel avenue for treatment of Her-2/neu-driven breast carcinomas.


Subject(s)
Enzyme Precursors/metabolism , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Protein-Lysine 6-Oxidase/metabolism , Receptor, ErbB-2/metabolism , Animals , Cell Adhesion/physiology , Cell Growth Processes/physiology , Mice , NIH 3T3 Cells , Phenotype , Signal Transduction
6.
J Cell Physiol ; 203(1): 111-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15368541

ABSTRACT

The deposition of insoluble functional collagen occurs following extracellular proteolytic processing of procollagens by procollagen N- and C-proteinases, fibril formation, and lysyl oxidase dependent cross-linking. Procollagen C-proteinases in addition process and activate lysyl oxidase. The present study evaluates a possible role for procollagen C-proteinases in controlling different aspects of collagen deposition in vitro. Studies determine whether inhibition of procollagen C-proteinase activity with a specific BMP-1 inhibitor results in perturbations in lysyl oxidase activation, and in collagen processing, deposition, and cross-linking in phenotypically normal cultured murine MC3T3-E1 cells. Data show that BMP-1 Inhibitor dose dependently inhibits lysyl oxidase activation by up to 50% in undifferentiated proliferating cells. In differentiating cultures, BMP-1 inhibitor decreased collagen processing but did not inhibit the accumulation of mature collagen cross-links. Finally, electron microscopy studies show that collagen fibril diameter increased. Thus, inhibition of procollagen C-proteinases results in perturbed collagen deposition primarily via decreased collagen processing.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Collagen Type I/metabolism , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/metabolism , Osteoblasts/enzymology , Protein-Lysine 6-Oxidase/metabolism , Animals , Bone Morphogenetic Protein 1 , Cell Differentiation , Cell Division , Cells, Cultured , Cross-Linking Reagents/metabolism , Drug Interactions , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacology , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Protein Processing, Post-Translational , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
7.
J Biol Chem ; 279(39): 40593-600, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15277520

ABSTRACT

Lysyl oxidase is an extracellular enzyme critical for the normal biosynthesis of collagens and elastin. In addition, lysyl oxidase reverts ras-mediated transformation, and lysyl oxidase expression is down-regulated in human cancers. Since suramin inhibits growth factor signaling pathways and induces lysyl oxidase in ras-transformed NIH3T3 cells (RS485 cells), we sought to investigate the effects of suramin on the phenotype of transformed cells and the role of lysyl oxidase in mediating these effects. Suramin treatment resulted in a more normal phenotype as judged by growth rate, cell cycle parameters, and morphology. beta-aminopropionitrile, the selective inhibitor of lysyl oxidase enzyme activity, was remarkably unable to block suramin-induced reversion. By contrast, ectopic antisense lysyl oxidase demonstrated that lysyl oxidase gene expression mediated phenotypic reversion. Since lysyl oxidase is synthesized as a 50 kDa precursor and processed to a 30 kDa active enzyme and 18 kDa propeptide, the effects of these two products on the transformed phenotype of RS485 cells were then directly assessed in the absence of suramin. Here we report, for the first time, that the lysyl oxidase propeptide, and not the lysyl oxidase enzyme, inhibits ras-dependent transformation as determined by effects on cell proliferation assays, growth in soft agar, and Akt-dependent induction of NF-kappaB activity. Thus, the lysyl oxidase propeptide, which is released during extracellular proteolytic processing of pro-lysyl oxidase, functions to inhibit ras-dependent cell transformation.


Subject(s)
Protein-Lysine 6-Oxidase/chemistry , Aminopropionitrile/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Cycle , Cell Division , Cell Line, Transformed , Collagen/chemistry , Elastin/chemistry , Genetic Vectors , Luciferases/metabolism , Mice , Microscopy, Fluorescence , NF-kappa B/metabolism , NIH 3T3 Cells , Oligonucleotides, Antisense/chemistry , Peptides/chemistry , Phenotype , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein-Lysine 6-Oxidase/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/metabolism , Signal Transduction , Suramin/pharmacology , Time Factors , Transfection
8.
J Cell Physiol ; 200(1): 53-62, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15137057

ABSTRACT

Differentiation of phenotypically normal osteoblast cultures leads to formation of a bone-like extracellular matrix in vitro. Maximum collagen synthesis occurs early in the life of these cultures, whereas insoluble collagen deposition occurs later and is accompanied by a diminished rate of collagen synthesis. The mechanisms that control collagen deposition seem likely to include regulation of extracellular collagen biosynthetic enzymes, but expression patterns of these enzymes in differentiating osteoblasts has received little attention. The present study determined the regulation of lysyl oxidase as a function of differentiation of phenotypically normal murine MC3T3-E1 cells at the level of RNA and protein expression and enzyme activity. In addition, the regulation of BMP-1/mTLD mRNA levels that encodes procollagen C-proteinases was assayed. The role of lysyl oxidase in controlling insoluble collagen accumulation was further investigated in inhibition studies utilizing beta-aminopropionitrile, a specific inhibitor of lysyl oxidase enzyme activity. Results indicate that lysyl oxidase is regulated as a function of differentiation of MC3T3-E1 cells, and that the maximum increase in lysyl oxidase activity precedes the most efficient phase of insoluble collagen accumulation. By contrast BMP-1/mTLD is more constitutively expressed. Inhibition of lysyl oxidase in these cultures increases the accumulation of abnormal collagen fibrils, as determined by solubility studies and by electron microscopy. Taken together, these data support that regulation of lysyl oxidase activity plays a key role in the control of collagen deposition by osteoblast cultures.


Subject(s)
Cell Differentiation , Collagen/metabolism , Gene Expression Regulation, Enzymologic , Osteoblasts/metabolism , Protein-Lysine 6-Oxidase/metabolism , Alkaline Phosphatase/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Line , Collagen/ultrastructure , Immunohistochemistry , Kinetics , Mice , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoblasts/ultrastructure , Protein-Lysine 6-Oxidase/genetics , Skull/cytology , Solubility
9.
J Biol Chem ; 279(29): 30060-5, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15138266

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) inhibits osteoblast function in vitro by inhibiting collagen deposition. Studies generally support that TNF-alpha does not inhibit collagen biosynthesis by osteoblasts but that collagen deposition is in some way diminished. The study investigated TNF-alpha regulation of biosynthetic enzymes and proteins crucial for posttranslational extracellular collagen maturation in osteoblasts including procollagen C-proteinases, procollagen C-proteinase enhancer, and lysyl oxidase. The working hypothesis is that such regulation could inhibit collagen deposition by osteoblasts. We report that in phenotypically normal MC3T3-E1 osteoblasts, TNF-alpha decreases collagen deposition without decreasing collagen mRNA levels or procollagen protein synthesis. Analyses of the cell layers revealed that TNF-alpha diminished the levels of mature collagen cross-links, pyridinoline and deoxypyridinoline. Further analyses revealed that the mRNA expression for lysyl oxidase, the determining enzyme required for collagen cross-linking, is down-regulated by TNF-alpha in a concentration- and time-dependent manner by up to 50%. The decrease was accompanied by a significant reduction of lysyl oxidase protein levels and enzyme activity. By contrast, Northern and Western blotting studies revealed that procollagen C-proteinases bone morphogenic protein-1 and mammalians Tolloid and procollagen C-proteinase enhancer were expressed in MC3T3-E1 cells and not down-regulated. The data together demonstrate that TNF-alpha does not inhibit collagen synthesis but does inhibit the expression and activity of lysyl oxidase in osteoblasts, thereby contributing to perturbed collagen cross-linking and accumulation. These studies identify a novel mechanism in which proinflammatory cytokine modulation of an extracellular biosynthetic enzyme plays a determining role in the control of collagen accumulation by osteoblasts.


Subject(s)
Collagen/metabolism , Osteoblasts/metabolism , Protein-Lysine 6-Oxidase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acids/pharmacology , Animals , Blotting, Northern , Blotting, Western , Cell Line , Cross-Linking Reagents/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Inflammation , Mice , Protein Structure, Tertiary , RNA, Messenger/metabolism , Time Factors
10.
J Biol Chem ; 278(33): 30781-7, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12788924

ABSTRACT

Lysyl oxidase catalyzes oxidative deamination of peptidyl-lysine and hydroxylysine residues in collagens and lysine residues in elastin to form peptidyl aldehydes that are required for the formation of covalent cross-links in normal extracellular matrix biosynthesis. Lysyl oxidase in addition has tumor suppressor activity, and phenotypic reversion of transformed cell lines is accompanied by increased lysyl oxidase expression. The mechanism of low expression of lysyl oxidase in tumor cells is unknown. The present study investigates the hypothesis that autocrine growth factor pathways maintain low lysyl oxidase expression levels in c-H-ras-transformed fibroblasts (RS485 cell line). Autocrine pathways were blocked with suramin, a general inhibitor of growth factor receptor binding, and resulted in more than a 10-fold increase in lysyl oxidase expression and proenzyme production. This regulation was found to be reversible and occurred at the transcriptional level determined using lysyl oxidase promoter/reporter gene assays. Function blocking anti-fibroblast growth factor-2 (FGF-2) antibody enhanced lysyl oxidase expression in the absence of suramin. Finally, the addition of FGF-2 to suramin-treated cells completely reversed suramin stimulation of lysyl oxidase mRNA levels. Data support that an FGF-2 autocrine pathway inhibits lysyl oxidase transcription in the tumorigenic-transformed RS485 cell line. This finding may be of therapeutic significance and, in addition, provides a new experimental approach to investigate the mechanism of the tumor suppressor activity of lysyl oxidase.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Protein-Lysine 6-Oxidase/genetics , 3T3 Cells , Animals , Antineoplastic Agents/pharmacology , Autocrine Communication/drug effects , Cell Line, Transformed , Dose-Response Relationship, Drug , Drug Interactions , Genes, Reporter , Genes, ras , Luciferases , Mice , Promoter Regions, Genetic , RNA, Messenger/analysis , Suramin/pharmacology , Up-Regulation/drug effects
11.
Anal Biochem ; 300(2): 245-51, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11779117

ABSTRACT

Lysyl oxidase catalyzes the final known enzymatic step required for collagen and elastin cross-linking in the biosynthesis of normal mature functional insoluble extracellular matrices. In addition, lysyl oxidase has been identified as a possible tumor suppressor. Lysyl oxidase activity in biological samples is traditionally and most reliably assessed by tritium release end-point assays using radiolabeled collagen or elastin substrates involving laborious vacuum distillation of the released tritiated water. In addition, a less sensitive fluorometric method exists that employs nonpeptidyl amine lysyl oxidase substrates and measures hydrogen peroxide production with horseradish peroxidase coupled to homovanillate oxidation. The present study describes a more sensitive fluorescent assay for lysyl oxidase activity that utilizes 1,5-diaminopentane as substrate, and released hydrogen peroxide is detected using Amplex red in horseradish peroxidase-coupled reactions. This method allows the detection of 40 ng of enzyme per 2 ml assay at 37 degrees C and is 7.5 times more sensitive than the currently available fluorometric assay for enzyme activity. This method eliminates the interference that occurs in some biological samples and can be successfully used to detect lysyl oxidase activity in cell culture experiments.


Subject(s)
Fluorometry/methods , Protein-Lysine 6-Oxidase/analysis , Protein-Lysine 6-Oxidase/metabolism , 3T3 Cells , Animals , Aorta/enzymology , Cattle , Cells, Cultured , Chromogenic Compounds/metabolism , Homovanillic Acid/metabolism , Hydrogen Peroxide/metabolism , Kinetics , Mice , Oxazines/metabolism , Sensitivity and Specificity , Tritium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL