Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Protein Expr Purif ; 131: 7-15, 2017 03.
Article in English | MEDLINE | ID: mdl-27815133

ABSTRACT

The aminopeptidase A (PepA; EC 3.4.11.7) is an intracellular exopeptidase present in lactic acid bacteria. The PepA cleaves glutamyl/aspartyl residues from the N-terminal end of peptides and can, therefore, be applied for the production of protein hydrolysates with an increased amount of these amino acids, which results in a savory taste (umami). The first PepA from a lactobacilli strain was recombinantly expressed in Escherichia coli in a recently published study and harbored a C-terminal His6-tag for easier purification. Due to the fact that a His-tag might influence the properties of an enzyme, a simple purification method for the non-His-tagged PepA was required. Surprisingly, the PepA precipitated at a very low ammonium sulfate concentration of 5%. Unusual for a precipitating step, the purity of PepA was over 95% and the obtained activity yield was 110%. The high purity allows biochemical characterization and kinetic investigation. As a result, the optimum pH (6.0-6.5) and temperature (60-65 °C) were comparable to the His6-tag harboring PepA; the KM value was at 0.79 mM slightly lower compared to 1.21 mM, respectively. Since PepA is a homo dodecamer, it has a high molecular mass of approximately 480 kDa. Therefore, a subsequent preparative size-exclusion chromatography (SEC) step seemed promising. The PepA after SEC was purified to homogeneity. In summary, the simple two-step purification method presented can be applied to purify high amounts of PepA that will allow the performance of experiments in the future to crystalize PepA for the first time.


Subject(s)
Bacterial Proteins , Glutamyl Aminopeptidase , Lactobacillus delbrueckii/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Enzyme Stability , Glutamyl Aminopeptidase/biosynthesis , Glutamyl Aminopeptidase/chemistry , Glutamyl Aminopeptidase/genetics , Glutamyl Aminopeptidase/isolation & purification , Hot Temperature , Hydrogen-Ion Concentration , Lactobacillus delbrueckii/enzymology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
2.
BMC Cancer ; 14: 386, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24885240

ABSTRACT

BACKGROUND: Advances in the knowledge of renal neoplasms have demonstrated the implication of several proteases in their genesis, growth and dissemination. Glutamyl-aminopeptidase (GAP) (EC. 3.4.11.7) is a zinc metallopeptidase with angiotensinase activity highly expressed in kidney tissues and its expression and activity have been associated wtih tumour development. METHODS: In this prospective study, GAP spectrofluorometric activity and immunohistochemical expression were analysed in clear-cell (CCRCC), papillary (PRCC) and chromophobe (ChRCC) renal cell carcinomas, and in renal oncocytoma (RO). Data obtained in tumour tissue were compared with those from the surrounding uninvolved kidney tissue. In CCRCC, classic pathological parameters such as grade, stage and tumour size were stratified following GAP data and analyzed for 5-year survival. RESULTS: GAP activity in both the membrane-bound and soluble fractions was sharply decreased and its immunohistochemical expression showed mild staining in the four histological types of renal tumours. Soluble and membrane-bound GAP activities correlated with tumour grade and size in CCRCCs. CONCLUSIONS: This study suggests a role for GAP in the neoplastic development of renal tumours and provides additional data for considering the activity and expression of this enzyme of interest in the diagnosis and prognosis of renal neoplasms.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Renal Cell/genetics , Glutamyl Aminopeptidase/biosynthesis , Kidney Neoplasms/genetics , Adenoma, Oxyphilic/genetics , Adenoma, Oxyphilic/pathology , Aged , Angiotensins/genetics , Angiotensins/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/pathology , Endopeptidases/genetics , Female , Gene Expression Regulation, Neoplastic , Glutamyl Aminopeptidase/genetics , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis
3.
J Control Release ; 145(1): 66-73, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20346382

ABSTRACT

The therapeutic index of anti-cancer drugs is increased when encapsulating them in tumor-targeted liposomes. Liposome-entrapped doxorubicin (DXR), targeting the tumor vasculature marker, aminopeptidase N (APN), displayed enhanced anti-tumor effects and prolonged survival in human neuroblastoma (NB)-bearing mice. Here we exploited a peptide ligand of aminopeptidase A (APA), discovered by phage display technology for delivery of liposomal DXR to perivascular tumor cells. Immunohistochemistry, performed in NB-bearing mice, showed APA expression in the vascular wall of NB primary and metastatic lesions. APA-targeted peptides displayed specific binding to APA-transfected cells in vitro, and also accumulation in the tumor of NB-bearing mice. Consequently, novel, APA-targeted, DXR-liposomes were developed and in vivo proof-of-principle was established, alone and in combination with APN-targeted DXR-loaded liposomes, in NB-bearing mice. Mice receiving APA-targeted liposomal DXR exhibited an increased life span in comparison to control mice, but to a lesser extent relative to that in mice treated with APN-targeted formulation, moreover the greatest increase in TUNEL-positive tumor cells was observed in animals treated with APN-targeted formulations. Mice treated with a combination of APA- and APN-targeted, liposomal DXR had a significant increase in life span compared to each treatment administered separately. There was a significant increase in the level of apoptosis in the tumors of mice on the combination therapy, and a pronounced destruction of the tumor vasculature with nearly total ablation of endothelial cells and pericytes. The availability of novel ligands binding to additional tumor vasculature-associated antigens will allow the design of sophisticated combinations of ligand-targeted liposomal anti-cancer drugs.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/therapeutic use , Endothelial Cells/drug effects , Neuroblastoma/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , CD13 Antigens/biosynthesis , CD13 Antigens/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Compounding , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Glutamyl Aminopeptidase/biosynthesis , Glutamyl Aminopeptidase/chemistry , Humans , Immunohistochemistry , Ligands , Liposomes , Mice , Mice, Nude , Neuroblastoma/blood supply , Neuroblastoma/metabolism , Neuroblastoma/pathology , Peptides/chemistry , Peptides/metabolism , Protein Binding , Xenograft Model Antitumor Assays
4.
Neuroscience ; 164(3): 1263-73, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19747525

ABSTRACT

Endogenous angiotensin (Ang) II and/or an Ang II-derived peptide, acting on Ang type 1 (AT(1)) and Ang type 2 (AT(2)) receptors, can carry out part of the nociceptive control modulated by periaqueductal gray matter (PAG). However, neither the identity of this putative Ang-peptide, nor its relationship to Ang II antinociceptive activity was clarified. Therefore, we have used tail-flick and incision allodynia models combined with an HPLC time course of Ang metabolism, to study the Ang III antinociceptive effect in the rat ventrolateral (vl) PAG using peptidase inhibitors and receptor antagonists. Ang III injection into the vlPAG increased tail-flick latency, which was fully blocked by Losartan and CGP 42,112A, but not by divalinal-Ang IV, indicating that Ang III effect was mediated by AT(1) and AT(2) receptors, but not by the AT(4) receptor. Ang III injected into the vlPAG reduced incision allodynia. Incubation of Ang II with punches of vlPAG homogenate formed Ang III, Ang (1-7) and Ang IV. Amastatin (AM) inhibited the formation of Ang III from Ang II by homogenate, and blocked the antinociceptive activity of Ang II injection into vlPAG, suggesting that aminopeptidase A (APA) formed Ang III from Ang II. Ang III can also be formed from Ang I by a vlPAG alternative pathway. Therefore, the present work shows, for the first time, that: (i) Ang III, acting on AT(1) and AT(2) receptors, can elicit vlPAG-mediated antinociception, (ii) the conversion of Ang II to Ang III in the vlPAG is required to elicit antinociception, and (iii) the antinociceptive activity of endogenous Ang II in vlPAG can be ascribed preponderantly to Ang III.


Subject(s)
Analgesics/pharmacology , Angiotensin III/metabolism , Nociceptors/drug effects , Pain/metabolism , Periaqueductal Gray/metabolism , Analgesics/metabolism , Angiotensin II/pharmacology , Angiotensin III/pharmacology , Angiotensin Receptor Antagonists , Animals , Disease Models, Animal , Drug Interactions/physiology , Efferent Pathways/drug effects , Efferent Pathways/metabolism , Glutamyl Aminopeptidase/biosynthesis , Losartan/pharmacology , Male , Microinjections , Neural Inhibition/drug effects , Neural Inhibition/physiology , Nociceptors/metabolism , Oligopeptides/pharmacology , Pain/drug therapy , Pain/physiopathology , Pain Measurement , Pain Threshold/drug effects , Pain Threshold/physiology , Peptides/pharmacology , Periaqueductal Gray/drug effects , Rats , Rats, Wistar , Receptors, Angiotensin/metabolism
5.
Cell Biol Int ; 32(7): 748-53, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18387830

ABSTRACT

Glutamyl aminopeptidase (GluAP, EC 3.4.11.7, ENPEP) is a 130-kDa homodimeric zinc metallopeptidase which specifically cleaves the N-terminal glutamate or aspartate residue of peptidic substrates such as cholecystokinin-8 or angiotensin (Ang) II, in vitro. We used a DNA microarray hybridization (Genechip Rat Expression Array 230A, Affymetrix Inc., Santa Clara, CA, USA) to demonstrate that GluAP was upregulated in osteogenic induced rat bone marrow stromal cells (BMSCs). To compare the expression of GluAP in the osteogenic differentiation and non-osteogenic differentiation of rat BMSCs in vitro, the cells were osteogenic induced in vitro. We also performed an MTT assay, alkaline phosphatase assay, alizarin red staining, and an immunohistochemical analysis to determine the osteogenic differentiation of BMSCs. The expression of GluAP was examined by real-time polymerase chain reaction (PCR). The real-time PCR results showed that GluAP was upregulated in osteogenic differentiated BMSCs in vitro, suggesting that GluAP may be correlated with the osteogenic differentiation of BMSCs.


Subject(s)
Bone Marrow Cells/metabolism , Glutamyl Aminopeptidase/biosynthesis , Stromal Cells/metabolism , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Proliferation , Collagen/metabolism , Gene Expression , Glutamyl Aminopeptidase/genetics , Male , Oligonucleotide Array Sequence Analysis , Osteogenesis , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Stromal Cells/cytology , Up-Regulation
6.
Am J Physiol Renal Physiol ; 292(2): F780-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16985214

ABSTRACT

Peptides play important roles in cell regulation and signaling in many tissues and are regulated by peptidases, most of which are highly expressed in the kidney. Several peptide convertases have a function in different tumor stages, and some have been clearly characterized as diagnostic and prognostic markers for solid tumors, including renal cancer; however, little is known about their in vivo role in kidney tumors. The present study compares the activity of a range of peptidases in human tumor samples and nontumor tissue obtained from clear cell renal cell carcinoma (CCRCC) patients. To cover the complete spectrum and subcellular distribution of peptide-converting activity, acid, neutral, basic, and omega activities were selected. CCRCC displays a selective and restricted pattern of peptidase activities. Puromycin-sensitive aminopeptidase activity in the tumor increases [tumor (t) = 10,775 vs. nontumor (n) = 7,635 units of peptidase (UP)/mg protein; P < 0.05], whereas aminopeptidase N decreases (t = 6,664 vs. n = 33,381 UP/mg protein; P < 0.001). Aminopeptidase B activity of the particulate fraction in tumors decreases (t = 2,399 vs. n = 13,536 UP/mg protein; P < 0.001) compared with nontumor tissues, and aspartyl-aminopeptidase activity decreases significantly in CCRCC (t = 137 vs. n = 223 UP/mg protein; P < 0.05). Soluble and particulate pyroglutamyl peptidase I activities, aminopeptidase A activity, and soluble aminopeptidase B activity do not vary in renal cancer. The relative expression for the aforementioned peptidases, assayed using quantitative RT-PCR, increases in CCRCC for aminopeptidases B (1.5-fold) and A (19-fold), aspartyl-aminopeptidase (3.9-fold), puromycin-sensitive aminopeptidase (2.5-fold), and pyroglutamyl peptidase I (7.6-fold). Only aminopeptidase N expression decreases in tumors (1.3-fold). This peptidase activity profile in the neoplastic kidney suggests a specific role for the studied convertases and the possible involvement of an intracrine renin-angiotensin system in the pathogenesis of CCRCC.


Subject(s)
Carcinoma, Renal Cell/enzymology , Kidney Neoplasms/enzymology , Peptide Hydrolases/metabolism , Adult , Aged , Aminopeptidases/biosynthesis , CD13 Antigens/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Glutamyl Aminopeptidase/biosynthesis , Humans , Kidney/enzymology , Male , Middle Aged , Pyroglutamyl-Peptidase I/biosynthesis
7.
J Histochem Cytochem ; 54(2): 253-62, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16286663

ABSTRACT

Aminopeptidase-A (APA) is a metalloprotease that cleaves N-terminal aspartyl and glutamyl residues from peptides. Its best-known substrate is angiotensin II (Ang II), the most active compound of the renin-angiotensin system (RAS). The RAS is involved in renal development. Most components of the RAS system are expressed in the developing kidney. Thus far, APA has not been studied in detail. In the present study we have evaluated the expression of APA at the protein, mRNA, and enzyme activity (EA) level in the kidney during nephrogenesis. Furthermore, we have studied the effect of inhibiting APA EA by injection of anti-APA antibodies into 1-day-old mice. APA expression was observed from the comma stage onwards, predominantly in the developing podocytes and brush borders of proximal tubular cells. Notably, APA was absent in the medulla or the renal arterioles. Inhibition of APA EA caused temporary podocyte foot-process effacement, suggesting a minimum role for APA during nephrogenesis.


Subject(s)
Glutamyl Aminopeptidase/antagonists & inhibitors , Glutamyl Aminopeptidase/biosynthesis , Kidney/enzymology , Animals , Animals, Newborn , Antibodies, Monoclonal/pharmacology , Glutamyl Aminopeptidase/genetics , Kidney/embryology , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/ultrastructure , Mice , Mice, Inbred BALB C , Microvilli/enzymology , Podocytes/enzymology , RNA, Messenger/biosynthesis
8.
Kidney Int ; 61(1 Suppl): S106-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11841622

ABSTRACT

BACKGROUND: High protein diets and diabetes increase renal renin angiotensin system (RAS) activity, which is associated with glomerular injury. Aminopeptidase A (APA) is a cell surface metalloprotease that degrades angiotensin II (AII) in the mesangium. Mesangial cells (MC) also possess receptors for AII; the type 1 (AT1 receptor) promotes proliferation and fibrosis, while the type 2 (AT2 receptor) opposes these effects. We evaluated whether amino acids and glucose alter expression of APA, AT1 receptor and AT2 receptor in a manner that further augments RAS activity. METHODS: Confluent rat MC were grown in serum-free media for 48 hours prior to exposing to experimental conditions: control (C), high amino acids (HA, mixed amino acid solution added to raise concentrations 5- to 6-fold over C), high glucose (HG 30, mM glucose). Semi-quantitative RT-PCR was used to assess mRNA for APA, AT1 receptor, AT2 receptor, and beta-actin. Values are expressed relative to beta actin. RESULTS: Both HA and HG reduced APA mRNA (HG 1.13 plus minus 0.19, HA 1.12 plus minus 0.16 versus C 1.27 plus minus 0.16 P < 0.05, N = 8). HA increased AT1 receptor mRNA (HA 2.11 plus minus 0.43 versus C 1.14 plus minus 0.28 P < 0.05, N = 8). HG increased AT2 receptor mRNA (HG 1.31 plus minus 0.43 versus C 0.82 plus minus 0.33 P < 0.05, N = 6). CONCLUSIONS: A reduction of APA, in response to high levels of amino acids or glucose, could contribute to increased AII as a result of decreased degradation in MC. The effect of amino acids to increase AT1 receptor expression may further enhance adverse hemodynamic and pro-fibrotic actions of AII. Conversely, glucose increased AT2 receptor expression, which could modulate responses mediated by the AT1 receptor.


Subject(s)
Amino Acids/pharmacology , Glucose/pharmacology , Glutamyl Aminopeptidase/biosynthesis , Mesangial Cells/drug effects , Mesangial Cells/physiology , Receptor, Angiotensin, Type 1/biosynthesis , Receptor, Angiotensin, Type 2/biosynthesis , Animals , Cells, Cultured , Rats , Rats, Sprague-Dawley
9.
Int J Mol Med ; 2(4): 409-14, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9857225

ABSTRACT

The regulatory mechanisms responsible for malignant transformation, tumor progression and metastasis in renal cell cancer (RCC) are still unclear, but there is some evidence that biologically active peptides might have regulatory effects on the behavior of this malignancy. Tumor cells can change local concentrations of active peptides by modulating their cell-surface enzymes. Using immunohistochemistry and enzyme-histochemistry, the expression of various membrane peptidases was examined in RCC and adjacent noninvaded renal parenchyma (n = 44). We describe the down-regulation of neutral endopeptidase 24.11 (NEP) protein expression in RCC of the clear cell/chromophilic type when compared with renal parenchyma, and show for the first time the lack of enzyme activity of NEP in RCC. The strongest expression could be found for dipeptidyl peptidase IV (DPIV) which is only decreased in RCC of the chromophobe cell type and is even present in oncocytoma. Aminopeptidase N (APN) and aminopeptidase A (APA) show attenuated expression in up to one third of clear cell/ chromophilic RCC. Chromophobe RCC and oncocytomas do not express APN, APA, NEP and gamma-glutamyltranspeptidase.


Subject(s)
Carcinoma, Renal Cell/enzymology , Kidney Neoplasms/enzymology , Kidney/enzymology , Neprilysin/biosynthesis , Adenoma, Oxyphilic/enzymology , Adenoma, Oxyphilic/pathology , Adult , Aged , CD13 Antigens/biosynthesis , Carcinoma, Renal Cell/pathology , Cell Membrane/enzymology , Dipeptidyl Peptidase 4/biosynthesis , Down-Regulation , Female , Glutamyl Aminopeptidase/biosynthesis , Histocytochemistry , Humans , Kidney/pathology , Kidney Neoplasms/pathology , Male , Middle Aged , gamma-Glutamyltransferase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...