Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Methods Cell Biol ; 126: 63-83, 2015.
Article in English | MEDLINE | ID: mdl-25665441

ABSTRACT

Incorporation of radioactive precursors of amino acids and/or modifier groups into proteins, isolation and sizing of polypeptide species of interest, and finally their detection and characterization provide a robust handle to examine the life cycle and varied modifications of any protein. A prerequisite in application of these techniques to lysosomal enzymes is the availability of an avid and specific antibody, because lysosomal proteins represent a very minor fraction of the cellular protein and must be purified without a significant loss many 1000-fold as conveniently as possible. Pulse-chase labeling and good knowledge on organelle-specific modifications of lysosomal proteins may enhance the information that can be obtained from such experiments. We describe procedures for pulse-chase labeling experiments that have proven to work with a commercially available antibody against a mouse and a human lysosomal protease and can be used as a reference in establishing the technique in any laboratory that has an access to a certified isotope facility and the knowledge to handle radioactivity safely. We discuss the crucial steps and refer to alternatives described in the literature. The present model protein cathepsin Z is synthesized as a larger proenzyme that contains two N-linked oligosaccharides and matures to a shorter single chain enzyme retaining the processed oligosaccharides. A pulse-chase experiment demonstrates the conversion of the precursor into the mature form. In addition, results on deglycosylation of metabolically labeled cathepsin Z are shown and the alterations in the apparent size of the glycopeptides are explained.


Subject(s)
Lysosomes/metabolism , Animals , Cathepsin Z/isolation & purification , Cathepsin Z/metabolism , Cells, Cultured , Humans , Immunoprecipitation , Protein Biosynthesis , Protein Processing, Post-Translational , Protein Transport , Proteins/isolation & purification , Proteins/metabolism , Staining and Labeling
2.
Biochimie ; 94(3): 684-94, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21978926

ABSTRACT

Growing evidence suggests the presence of active lysosomal enzymes in extra-lysosomal compartments, such as the plasma membrane. Although in the past little attention was paid to glycohydrolases acting on cellular compartments different from lysosomes, there is now increasing interest on plasma membrane-associated glycohydrolases because they should be involved, together with glycosyltransferases, in glycosphingolipids oligosaccharide modification processes regulating cell-to-cell and/or cell-environment interactions in both physiological and pathological conditions. Starting from the previous evidence of the presence of ß-hexosaminidase and ß-galactosidase at the plasma membrane of cultured fibroblasts, we here investigated the association of these glycohydrolases with lipid microdomains of Jurkat T-lymphocytes. Monosialoganglioside GM3 represents the major glycosphingolipid constituent of T-cell plasma membrane and its amount largely increases after T-cell stimulation. ß-hexosaminidase and ß-galactosidase cleave specific ß-linked terminal residues from a wide range of glycoconjugates and in particular are involved in the stepwise degradation of GM1 to GM3 ganglioside. Here we demonstrated that fully processed plasma membrane-associated ß-hexosaminidase and ß-galactosidase co-distribute with the lipid microdomain markers and co-immunoprecipitate with the signalling protein lck in Jurkat T-cell. Furthermore, Jurkat cell stimulation up-regulates the expression and activity of lysosomal ß-hexosaminidase and ß-galactosidase and increases their targeting to lipid microdomains. The non-random distribution of plasma membrane-associated ß-hexosaminidase and ß-galactosidase and their localization within lipid microdomains, suggest a role of these enzymes in the local reorganization of glycosphingolipid-based signalling units.


Subject(s)
Jurkat Cells/metabolism , Membrane Microdomains/metabolism , T-Lymphocytes/metabolism , beta-Galactosidase/metabolism , beta-N-Acetylhexosaminidases/metabolism , Cell Line , Humans , Real-Time Polymerase Chain Reaction , beta-Galactosidase/genetics , beta-N-Acetylhexosaminidases/genetics
3.
Biochem J ; 439(1): 113-28, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21692750

ABSTRACT

DIRC2 (Disrupted in renal carcinoma 2) has been initially identified as a breakpoint-spanning gene in a chromosomal translocation putatively associated with the development of renal cancer. The DIRC2 protein belongs to the MFS (major facilitator superfamily) and has been previously detected by organellar proteomics as a tentative constituent of lysosomal membranes. In the present study, lysosomal residence of overexpressed as well as endogenous DIRC2 was shown by several approaches. DIRC2 is proteolytically processed into a N-glycosylated N-terminal and a non-glycosylated C-terminal fragment respectively. Proteolytic cleavage occurs in lysosomal compartments and critically depends on the activity of cathepsin L which was found to be indispensable for this process in murine embryonic fibroblasts. The cleavage site within DIRC2 was mapped between amino acid residues 214 and 261 using internal epitope tags, and is presumably located within the tentative fifth intralysosomal loop, assuming the typical MFS topology. Lysosomal targeting of DIRC2 was demonstrated to be mediated by a N-terminal dileucine motif. By disrupting this motif, DIRC2 can be redirected to the plasma membrane. Finally, in a whole-cell electrophysiological assay based on heterologous expression of the targeting mutant at the plasma membrane of Xenopus oocytes, the application of a complex metabolic mixture evokes an outward current associated with the surface expression of full-length DIRC2. Taken together, these data strongly support the idea that DIRC2 is an electrogenic lysosomal metabolite transporter which is subjected to and presumably modulated by limited proteolytic processing.


Subject(s)
Cathepsin L/metabolism , Lysosomal Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Cathepsin L/genetics , Computational Biology , Electrophysiology , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Lysosomal Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Mice , Neoplasm Proteins/genetics , Protein Binding , Xenopus
4.
Peptides ; 32(5): 881-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21396418

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, manifests discreet strategies to subvert host immune responses, which enable the pathogen to survive and multiply inside the macrophages. This problem is further worsened by the emergence of multidrug resistant mycobacterial strains, which make most of the anti-tuberculous drugs ineffective. It is thus imperative to search for and design better therapeutic strategies, including employment of new antibiotics. Recently, naturally produced antimicrobial molecules such as enzymes, peptides and their synthetic analogs have emerged as compounds with potentially significant therapeutical applications. Although, many antimicrobial peptides have been identified only very few of them have been tested against mycobacteria. A major limitation in using peptides as therapeutics is their sensitivity to enzymatic degradation or inactivity under certain physiological conditions such as relatively high salt concentration. Here, we show that NK-2, a peptide representing the cationic core region of the lymphocytic effector protein NK-lysin, and Ci-MAM-A24, a synthetic salt-tolerant peptide derived from immune cells of Ciona intestinalis, efficiently kill Mycobacterium smegmatis and Mycobacterium bovis-BCG. In addition, NK-2 and Ci-MAM-A24 showed a synergistic killing effect against M. smegmatis, no cytotoxic effect on mouse macrophages at bactericidal concentrations, and were even found to kill mycobacteria residing inside the macrophages. We also show that human placental lysosomal contents exert potent killing effect against mycobacteria under acidic and reducing growth conditions. Electron microscopic studies demonstrate that the lysosomal extract disintegrate bacterial cell membrane resulting in killing of mycobacteria.


Subject(s)
Anti-Infective Agents/pharmacology , Lysosomes/chemistry , Mycobacterium/drug effects , Peptides/pharmacology , Placental Extracts/pharmacology , 2,4-Dichlorophenoxyacetic Acid/analogs & derivatives , 2,4-Dichlorophenoxyacetic Acid/chemistry , 2,4-Dichlorophenoxyacetic Acid/pharmacology , Animals , Anti-Infective Agents/adverse effects , Anti-Infective Agents/chemistry , Cell Line , Drug Synergism , Humans , Macrophages/drug effects , Mice , Mycobacterium smegmatis/drug effects , Peptides/adverse effects , Peptides/chemistry
5.
Proteomics ; 10(22): 4053-76, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20957757

ABSTRACT

Lysosomes are organelles of eukaryotic cells that are critically involved in the degradation of macromolecules mainly delivered by endocytosis and autophagocytosis. Degradation is achieved by more than 60 hydrolases sequestered by a single phospholipid bilayer. The lysosomal membrane facilitates interaction and fusion with other compartments and harbours transport proteins catalysing the export of catabolites, thereby allowing their recycling. Lysosomal proteins have been addressed in various proteomic studies that are compared in this review regarding the source of material, the organelle/protein purification scheme, the proteomic methodology applied and the proteins identified. Distinguishing true constituents of an organelle from co-purifying contaminants is a central issue in subcellular proteomics, with additional implications for lysosomes as being the site of degradation of many cellular and extracellular proteins. Although many of the lysosomal hydrolases were identified by classical biochemical approaches, the knowledge about the protein composition of the lysosomal membrane has remained fragmentary for a long time. Using proteomics many novel lysosomal candidate proteins have been discovered and it can be expected that their functional characterisation will help to understand functions of lysosomes at a molecular level that have been characterised only phenomenologically so far and to generally deepen our understanding of this indispensable organelle.


Subject(s)
Lysosomes/chemistry , Proteome , Animals , Humans , Lipofuscin , Lysosomes/genetics , Lysosomes/physiology , Membrane Proteins , Mice , Proteomics , Rats
6.
Biol Chem ; 391(6): 695-704, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20370317

ABSTRACT

Transmembrane protein 192 (TMEM192) has been previously identified in proteomic analyses of lysosomal membranes. TMEM192 does not exhibit any significant homology to known protein families and possesses four potential transmembrane segments. To approach the molecular role of TMEM192, a detailed biochemical characterisation of this protein was performed. Expression constructs of fusion proteins containing TMEM192 and appended epitope tags were constructed. In HeLa cells these proteins were detected in membranes of lysosomes/late endosomes. To examine endogenous TMEM192, a TMEM192-specific antibody was generated and validated. With this antibody colocalisation of endogenous TMEM192 with lysosomal and late endosomal markers was demonstrated. Using Percoll density gradient centrifugation and immunoblotting, co-sedimentation of major portions of both TMEM192 and the lysosomal proteins LAMP-2 and cathepsin D into high-density fractions was observed. Interestingly, in contrast to many other lysosomal proteins no N-glycosylation of TMEM192 could be detected. Western blotting of reduced and non-reduced samples and co-immunoprecipitation experiments indicated TMEM192 to be a homodimer with one or more interchain disulphide bridges. TMEM192 was found to be strongly expressed in human kidney, liver, lung and pancreas tissue. The widespread tissue distribution could suggest an important role of TMEM192 for lysosomal function.


Subject(s)
Intracellular Membranes/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Computational Biology , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Membrane Proteins/chemistry , Proteomics
7.
Biochem J ; 422(1): 83-90, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19489740

ABSTRACT

Until recently, a modest number of approx. 40 lysosomal membrane proteins had been identified and even fewer were characterized in their function. In a proteomic study, using lysosomal membranes from human placenta we identified several candidate lysosomal membrane proteins and proved the lysosomal localization of two of them. In the present study, we demonstrate the lysosomal localization of the mouse orthologue of the human C1orf85 protein, which has been termed kidney-predominant protein NCU-G1 (GenBank accession number: AB027141). NCU-G1 encodes a 404 amino acid protein with a calculated molecular mass of 39 kDa. The bioinformatics analysis of its amino acid sequence suggests it is a type I transmembrane protein containing a single tyrosine-based consensus lysosomal sorting motif at position 400 within the 12-residue C-terminal tail. Its lysosomal localization was confirmed using immunofluorescence with a C-terminally His-tagged NCU-G1 and the lysosomal marker LAMP-1 (lysosome-associated membrane protein-1) as a reference, and by subcellular fractionation of mouse liver after a tyloxapol-induced density shift of the lysosomal fraction using an anti-NCU-G1 antiserum. In transiently transfected HT1080 and HeLa cells, the His-tagged NCU-G1 was detected in two molecular forms with apparent protein sizes of 70 and 80 kDa, and in mouse liver the endogenous wild-type NCU-G1 was detected as a 75 kDa protein. The remarkable difference between the apparent and the calculated molecular masses of NCU-G1 was shown, by digesting the protein with N-glycosidase F, to be due to an extensive glycosylation. The lysosomal localization was impaired by mutational replacement of an alanine residue for the tyrosine residue within the putative sorting motif.


Subject(s)
Lysosomes/metabolism , Membrane Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Centrifugation, Density Gradient , Cloning, Molecular , Computational Biology , Fluorescent Antibody Technique , Gene Expression Profiling , Glycosylation/drug effects , HeLa Cells , Humans , Lysosomes/drug effects , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Mutant Proteins/metabolism , Polyethylene Glycols/pharmacology , Protein Transport/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics
8.
Microbes Infect ; 11(10-11): 868-75, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19460455

ABSTRACT

Non-pathogenic mycobacteria such us Mycobacterium smegmatis reside in macrophages within phagosomes that fuse with late endocytic/lysosomal compartments. This sequential fusion process is required for the killing of non-pathogenic mycobacteria by macrophages. Porins are proteins that allow the influx of hydrophilic molecules across the mycobacterial outer membrane. Deletion of the porins MspA, MspC and MspD significantly increased survival of M. smegmatis in J774 macrophages. However, the mechanism underlying this observation is unknown. Internalization of wild-type M. smegmatis (SMR5) and the porin triple mutant (ML16) by macrophages was identical indicating that the viability of the porin mutant in vivo was enhanced. This was not due to effects on phagosome trafficking since fusion of phagosomes containing the mutant with late endocytic compartments was unaffected. Moreover, in ML16-infected macrophages, the generation of nitric oxide (NO) was similar to the wild type-infected cells. However, ML16 was significantly more resistant to the effects of NO in vitro compared to SMR5. Our data provide evidence that porins render mycobacteria vulnerable to killing by reactive nitrogen intermediates within phagosomes probably by facilitating uptake of NO across the mycobacterial outer membrane.


Subject(s)
Anti-Bacterial Agents/pharmacology , Macrophages/immunology , Macrophages/microbiology , Microbial Viability/drug effects , Mycobacterium smegmatis/drug effects , Nitric Oxide/pharmacology , Porins/physiology , Animals , Anti-Bacterial Agents/immunology , Cell Line , Colony Count, Microbial , Gene Deletion , Humans , Mice , Microbial Sensitivity Tests , Mycobacterium smegmatis/immunology , Nitric Oxide/immunology , Porins/genetics
9.
J Biol Chem ; 284(14): 9372-81, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19224915

ABSTRACT

Arylsulfatase A (ASA) catalyzes the intralysosomal desulfation of 3-O-sulfogalactosylceramide (sulfatide) to galactosylceramide. The reaction requires saposin B (Sap B), a non-enzymatic proteinaceous cofactor which presents sulfatide to the catalytic site of ASA. The lack of either ASA or Sap B results in a block of sulfatide degradation, progressive intralysosomal accumulation of sulfatide, and the fatal lysosomal storage disease metachromatic leukodystrophy. We studied the coupled Sap B-ASA reaction in vitro using detergent-free micellar and liposomal assay systems and in vivo using cell culture models of metachromatic leukodystrophy. Under in vitro conditions, the reaction had a narrow pH optimum around pH 4.3 and was inhibited by mono- and divalent cations, phosphate and sulfite. Bis(monoacylglycero) phosphate and phosphatidic acid were activators of the reaction, underscoring a significant role of acidic phosphoglycerolipids in sphingolipid degradation. Desulfation was negligible when Sap B was substituted by Sap A, C, or D. Up to a molar ratio between Sap B and sulfatide of 1:5, an elevation of Sap B concentrations caused a sharp increase of sulfatide hydrolysis, indicating the requirement of unexpected high Sap B levels for maximum turnover. Feeding of ASA-deficient, sulfatide-storing primary mouse kidney cells with ASA caused partial clearance of sulfatide. Co-feeding of Sap B or its precursor prosaposin resulted in the lysosomal uptake of the cofactor but did not promote ASA-catalyzed sulfatide hydrolysis. This suggests that Sap B is not a limiting factor of the coupled Sap B-ASA reaction in mouse kidney cells even if sulfatide has accumulated to unphysiologically high levels.


Subject(s)
Cerebroside-Sulfatase/metabolism , Leukodystrophy, Metachromatic/enzymology , Models, Biological , Saposins/metabolism , Animals , Cells, Cultured , Cerebroside-Sulfatase/deficiency , Cerebroside-Sulfatase/genetics , Disulfides/metabolism , Enzyme Activation , Humans , Hydrolysis , Leukodystrophy, Metachromatic/genetics , Lipid Metabolism , Liposomes , Mice , Mice, Knockout , Substrate Specificity , Swine
10.
Biosci Rep ; 28(4): 229-37, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18588514

ABSTRACT

Hex (beta-hexosaminidase) is a soluble glycohydrolase involved in glycoconjugate degradation in lysosomes, however its localization has also been described in the cytosol and PM (plasma membrane). We previously demonstrated that Hex associated with human fibroblast PM as the mature form, which is functionally active towards G(M2) ganglioside. In the present study, Hex was analysed in a lysosomal membrane-enriched fraction obtained by purification from highly purified human placenta lysosomes. These results demonstrate the presence of mature Hex associated with the lysosomal membrane and displaying, as observed for the PM-associated form, an acidic optimum pH. When subjected to sodium carbonate extraction, the enzyme behaved as a peripheral membrane protein, whereas Triton X-114 phase separation confirmed its partially hydrophilic nature, characteristics which are shared with the PM-associated form of Hex. Moreover, two-dimensional electrophoresis indicated a slight difference in the pI of beta-subunits in the membrane and the soluble forms of the lysosomal Hex. These results reveal a new aspect of Hex biology and suggest that a fully processed membrane-associated form of Hex is translocated from the lysosomal membrane to the PM by an as yet unknown mechanism. We present a testable hypothesis that, at the cell surface, Hex changes the composition of glycoconjugates that are known to be involved in intercellular communication and signalling.


Subject(s)
Lysosomes/enzymology , Placenta/enzymology , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/isolation & purification , beta-N-Acetylhexosaminidases/metabolism , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Hexosaminidase A/chemistry , Hexosaminidase A/isolation & purification , Hexosaminidase A/metabolism , Hexosaminidase B/chemistry , Hexosaminidase B/isolation & purification , Hexosaminidase B/metabolism , Humans , Intracellular Membranes/enzymology , Intracellular Membranes/metabolism , Lysosomes/metabolism , Placenta/metabolism
11.
Neurobiol Aging ; 29(1): 12-22, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17049675

ABSTRACT

Cathepsin D (CTSD), a protease detectable in different cell types whose primary function is to degrade proteins by bulk proteolysis in lysosomes, has been suggested to be involved in Alzheimer's disease (AD). In fact, there is increasing evidence that disturbance of the normal balance and localization of cathepsins may contribute to neurodegeneration in AD [Nakanishi H. Neuronal and microglial cathepsins in aging and age-related diseases. Aging Res Rev 2003; 2(4):367-81]. Here, we provide evidence of an altered balance of CTSD in skin fibroblasts from patients affected either by sporadic or familial forms of AD. In particular, we demonstrate that CTSD is down regulated at both transcriptional and translational level and its processing is altered in AD fibroblasts. The oncogene Ras is involved in the regulation of CTSD, as high expression level of the constitutively active form of Ras in normal or AD fibroblasts induces CTSD down-regulation. p38 MAPK signalling pathway also appears to down-modulate CTSD level. Overall results reinforce the hypothesis that a lysosomal impairment may be involved in AD pathogenesis and can be detected not only in the CNS but also at a peripheral level.


Subject(s)
Alzheimer Disease/pathology , Cathepsin D/metabolism , Down-Regulation/physiology , Fibroblasts/enzymology , Adult , Cells, Cultured , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Humans , Male , Middle Aged , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/physiology
12.
J Biol Chem ; 282(52): 37836-43, 2007 Dec 28.
Article in English | MEDLINE | ID: mdl-17977821

ABSTRACT

The delivery of protein fragments to major histocompatibility complex (MHC)-loading compartments of professional antigen-presenting cells is essential in the adaptive immune response against pathogens. Apart from the crucial role of the transporter associated with antigen processing (TAP) for peptide loading of MHC class I molecules in the endoplasmic reticulum, TAP-independent translocation pathways have been proposed but not identified so far. Based on its overlapping substrate specificity with TAP, we herein investigated the ABC transporter ABCB9, also named TAP-like (TAPL). Remarkably, TAPL expression is strongly induced during differentiation of monocytes to dendritic cells and to macrophages. TAPL does not, however, restore MHC class I surface expression in TAP-deficient cells, demonstrating that TAPL alone or in combination with single TAP subunits does not form a functional transport complex required for peptide loading of MHC I in the endoplasmic reticulum. In fact, by using quantitative immunofluorescence and subcellular fractionation, TAPL was detected in the lysosomal compartment co-localizing with the lysosome-associated membrane protein LAMP-2. By in vitro assays, we demonstrate a TAPL-specific translocation of peptides into isolated lysosomes, which strictly requires ATP hydrolysis. These results suggest a mechanism by which antigenic peptides have access to the lysosomal compartment in professional antigen-presenting cells.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Dendritic Cells/cytology , Lysosomes/chemistry , Peptides/chemistry , Antigen Presentation , Antigen-Presenting Cells , Antigens/chemistry , Biological Transport , Cell Line, Tumor , Cloning, Molecular , Dendritic Cells/metabolism , HeLa Cells , Humans , Lipopolysaccharide Receptors/biosynthesis , Lysosomes/metabolism , Models, Biological , Monocytes/metabolism
13.
Traffic ; 8(12): 1676-1686, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17897319

ABSTRACT

We searched for novel proteins in lysosomal membranes, tentatively participating in molecular transport across the membrane and/or in interactions with other compartments. In membranes purified from placental lysosomes, we identified 58 proteins, known to reside at least partially in the lysosomal membrane. These included 17 polypeptides comprising or associated with the vacuolar adenosine triphosphatase. We report on additional 86 proteins that were significantly enriched in the lysosomal membrane fraction. Among these, 12 novel proteins of unknown functions were found. Three were orthologues of rat proteins that have been identified in tritosomes by Bagshaw RD et al. (A proteomic analysis of lysosomal integral membrane proteins reveals the diverse composition of the organelle. Mol Cell Proteomics 2005;4:133-143). Here, the proteins encoded by LOC201931 (FLJ38482) and LOC51622 (C7orf28A) were expressed with an appended fluorescent tag in HeLa cells and found to be present in lysosomal organelles. Among the lysosomally enriched proteins, also 16 enzymes and transporters were detected that had not been assigned to lysosomal membranes previously. Finally, our results identified a particular set of proteins with known functions in signaling and targeting to be at least partially associated with lysosomes.


Subject(s)
Cell Membrane/metabolism , Lysosomes/metabolism , Adenosine Triphosphatases/chemistry , Biological Transport , Chromatography, Liquid/methods , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Intracellular Membranes/metabolism , Mass Spectrometry/methods , Microscopy, Electron , Models, Biological , Placenta/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteomics/methods , Recombinant Fusion Proteins/chemistry , Signal Transduction
14.
Cell Immunol ; 246(1): 1-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17617393

ABSTRACT

Mutations in the neutrophil elastase (NE) gene have been postulated to interfere with normal intracellular trafficking of NE as an AP3-interacting membrane integrated protein and to cause severe congenital or cyclic neutropenia in humans. Here, we show that in U937 promonocytes NE is synthesized as a predominantly soluble proenzyme and is completely secreted in the presence of phorbol esters similarly to serglycin. Using chemical cross-linking NE is shown to be associated with serglycin as 34 kDa proenzyme in the trans-Golgi region of these cells indicating that it is delivered to lysosomes associated with serglycin.


Subject(s)
Leukocyte Elastase/metabolism , Lysosomes/enzymology , Proteoglycans/metabolism , Vesicular Transport Proteins/metabolism , Ammonium Chloride/pharmacology , Cross-Linking Reagents/pharmacology , Glycoproteins/metabolism , Humans , Leukocyte Elastase/analysis , Monocytes/drug effects , Monocytes/enzymology , Monocytes/ultrastructure , Phorbol Esters/pharmacology , Protein Transport , Proteoglycans/analysis , Solubility , Tunicamycin/pharmacology , U937 Cells , Vesicular Transport Proteins/analysis , trans-Golgi Network/enzymology
15.
J Cell Biochem ; 101(6): 1558-66, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17340625

ABSTRACT

In several reports cathepsin D has been implicated in apoptosis. In some systems the effects of agents considered to be mediated by cathepsin D were inhibited in the presence of pepstatin A, an inhibitor of the enzyme. In other studies the effect of a mutant cathepsin D deprived of activity was indistinguishable from that of the normal enzyme. Here we show that in human fibroblasts and in HeLa cells apoptosis can be induced by microinjecting into cytosol either mature cathepsin D or its inactive precursor procathepsin D. The microinjected precursor remains in the uncleaved form. These results confirm that the proapoptotic effect of cathepsin D in the cytosol is independent of its catalytic activity and suggest that the interaction of cathepsin D with the downstream effector does not involve the active site of the enzyme, since in the proenzyme the active site is masked by the prosequence.


Subject(s)
Apoptosis/physiology , Cathepsin D/metabolism , Fibroblasts/physiology , Protein Precursors/metabolism , Binding Sites , Caspase 3/metabolism , Caspase Inhibitors , Cell Shape , Cells, Cultured , Cytoplasm/metabolism , Fibroblasts/cytology , HeLa Cells , Humans , Microinjections , Pepstatins/metabolism , Protease Inhibitors/metabolism
16.
J Leukoc Biol ; 81(4): 1149-58, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17210618

ABSTRACT

To clarify the sorting mechanism of the lysosomal/granular proteoglycan serglycin, we treated human promonocytic U937 cells with p-nitrophenyl-beta-D-xyloside (PNP-xyl) and cycloheximide. In the absence of protein synthesis, the carbohydrate moiety of serglycin was synthesized as PNP-xyl-chondroitin sulfate (CS), and most of it was delivered to lysosomes and degraded. Further, an augmented lysosomal targeting of serglycin in the presence of tunicamycin suggested that a sorting/lectin receptor with multiple specificity was involved with an increased capacity for serglycin in the absence of N-glycosylation. Correspondingly, the cation-independent mannose 6-phosphate receptor (CI-MPR) and sortilin were observed to bind to immobilized CS. These receptors were eluted in the presence of 200-400 mM and 100-250 mM NaCl, respectively. After treating the cells with a cross-linking reagent, a portion of the sulfated proteoglycan was coimmunoprecipitated with the CI-MPR but not with sortilin. In the presence of phorbol ester, lysosomal targeting of serglycin and to a lesser extent, of cathepsin D was inhibited. We conclude that the CI-MPR participates in lysosomal and granular targeting of serglycin and basic proteins such as lysozyme associated with the proteoglycan in hematopoietic cells.


Subject(s)
Lysosomes/metabolism , Proteoglycans/metabolism , Receptor, IGF Type 2/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Proteins, Vesicular Transport , Biological Transport , Cations/chemistry , Chondroitin Sulfates/chemistry , Chromatography, Affinity , Cross-Linking Reagents/pharmacology , HL-60 Cells , Humans , Immunoprecipitation , Lysosomes/drug effects , Lysosomes/enzymology , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Biosynthesis , Receptor, IGF Type 2/physiology , Tetradecanoylphorbol Acetate/pharmacology , Tunicamycin/pharmacology , U937 Cells
17.
FEBS Lett ; 581(1): 102-8, 2007 Jan 09.
Article in English | MEDLINE | ID: mdl-17174955

ABSTRACT

A structural hallmark of lysosomes is heterogeneity of their contents. We describe a method for isolation of particulate materials from human placental lysosomes. After a methionine methyl ester-induced disruption of lysosomes and two density gradient centrifugations we obtained a homogeneous membrane fraction and another one enriched in particulate inclusions. The latter exhibited a yellow-brown coloration and contained bodies lacking a delimiting membrane, which were characterised by a granular pattern and high electron density. The lipofuscin-like inclusion materials were rich in tripeptidyl peptidase I, beta-glucuronidase, acid ceramidase and apolipoprotein D and contained proteins originating from diverse subcellular localisations. Here we show that human term placenta contains lipofuscin-like lysosomal inclusions, a phenomenon usually associated with senescence in postmitotic cells. These findings imply that a simple pelleting of a lysosomal lysate is not appropriate for the isolation of lysosomal membranes, as the inclusions tend to be sedimented with the membranes.


Subject(s)
Inclusion Bodies/chemistry , Lipofuscin/chemistry , Lysosomes/chemistry , Placenta/chemistry , Pregnancy Proteins/chemistry , Cellular Senescence/physiology , Female , Humans , Inclusion Bodies/enzymology , Intracellular Membranes/chemistry , Intracellular Membranes/enzymology , Lipofuscin/metabolism , Lysosomes/enzymology , Placenta/enzymology , Pregnancy , Pregnancy Proteins/metabolism , Tripeptidyl-Peptidase 1
18.
ChemMedChem ; 1(4): 445-57, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16892380

ABSTRACT

A synthetic concept is presented that allows the construction of peptide isostere libraries through polymer-supported C-acylation reactions. A phosphorane linker reagent is used as a carbanion equivalent; by employing MSNT as a coupling reagent, the C-acylation can be conducted without racemization. Diastereoselective reduction was effected with L-selectride. The reagent linker allows the preparation of a norstatine library with full variation of the isosteric positions including the P1 side chain that addresses the protease S1 pocket. Therefore, the concept was employed to investigate the P1 site specificity of peptide isostere inhibitors systematically. The S1 pocket of several aspartic proteases including plasmepsin II and cathepsin D was modeled and docked with approximately 500 amino acid side chains. Inspired by this virtual screen, a P1 site mutation library was designed, synthesized, and screened against three aspartic proteases (plasmepsin II, HIV protease, and cathepsin D). The potency of norstatine inhibitors was found to depend strongly on the P1 substituent. Large, hydrophobic residues such as biphenyl, 4-bromophenyl, and 4-nitrophenyl enhanced the inhibitory activity (IC50) by up to 70-fold against plasmepsin II. In addition, P1 variation introduced significant selectivity, as up to 9-fold greater activity was found against plasmepsin II relative to human cathepsin D. The active P1 site residues did not fit into the crystal structure; however, molecular dynamics simulation suggested a possible alternative binding mode.


Subject(s)
Indicators and Reagents/chemistry , Molecular Mimicry , Protease Inhibitors/chemistry , Aminocaproates/chemistry , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cathepsin D/antagonists & inhibitors , HIV Protease/drug effects , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Protozoan Proteins , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism
20.
Protein Expr Purif ; 45(1): 157-67, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16242956

ABSTRACT

We used a vaccinia virus expression system for the production of recombinant human cathepsin D (CD), a lysosomal protease implicated in various patho-physiological processes including cancer, neurodegeneration, and development. The recombinant protein was successfully expressed in various human and non-human cells. It was correctly synthesized as a glycosylated 53 kDa precursor (proCDrec) that reacted with a polyclonal antibody against residues 7-21 of the propeptide sequence. In contrast to the control, in cells infected with the recombinant virus proCDrec was largely secreted into the culture medium, although it contained high-mannose oligosaccharides with uncovered mannose-6-phosphate residues. Intracellular proCDrec was processed into the 48 kDa intermediate single-chain and the 31 plus 13 kDa double-chain forms, however, the processing was slower than in normal cells. A method based on Pepstatin A-affinity chromatography allowed to isolate the recombinant protein from the medium of infected cells. Based on its latency in activity assay at acid pH and on its reactivity with antibodies specific for the N-terminus, the purified protein was judged to be in the inactive precursor form. During incubation at acid pH the purified proCDrec underwent autocatalytic processing and acquired pepstatin A-sensitive enzyme activity, as expected for correctly folded proCD. Antiserum raised in rabbits against proCDrec specifically reacted with human, but not with mouse proCD under non-denaturing conditions. We conclude that our vaccinia virus-directed proCDrec displays structural and functional features resembling those of native human proCD. This system can therefore be exploited for the synthesis of large quantities of human proCD, allowing further studies on the structure and function of this interesting protein.


Subject(s)
Cathepsin D , Enzyme Precursors , Animals , Cathepsin D/drug effects , Cathepsin D/isolation & purification , Cathepsin D/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , DNA, Complementary/genetics , Enzyme Precursors/drug effects , Enzyme Precursors/isolation & purification , Enzyme Precursors/metabolism , Haplorhini , HeLa Cells , Humans , Immune Sera/pharmacology , Mice , Oligosaccharides/chemistry , Phosphorylation , Rats , Recombinant Proteins/drug effects , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sensitivity and Specificity , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...