Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
1.
Appl Microbiol Biotechnol ; 103(15): 6081-6095, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31175430

ABSTRACT

ß-Glucuronidase is a lysosomal enzyme and a molecular model of a class of therapeutics approved as enzyme replacement therapies for lysosomal storage diseases. Understanding the effect of bioreactor process variables on the production and quality of the biologics is critical for maintaining quality and efficacy of the biotherapeutics. Here, we have investigated the effect of three process variables, in a head-to-head comparison using a parallel bioreactor system (n = 8), namely 0.25 mM butyrate addition, a temperature shift (from 37 to 32 °C), and a pH shift (from 7.0 to 6.7) along with a control (pH 7, temperature 37 °C, and no additive) on the production and quality of human recombinant ß-glucuronidase (GUS) by a Chinese hamster ovary (CHO) cell line. The study was performed as two independent runs (2 bioreactors per treatment per run; n ≤ 4). Although statistically not significant, protein production slightly increased with either 0.25 mM butyrate addition (13%) or pH shift (7%), whereas temperature shift decreased production (12%, not significant). Further characterization of the purified GUS samples showed that purification selectively enriched the mannose-6-phosphate (M6P)-containing GUS protein. Noticeably, a variation observed for the critical quality attribute (CQA) of the enzyme, namely M6P content, decreased after purification, across treatment replicates and, more so, across different treatments. The dimer content in the purified samples was comparable (~25%), and no significant discrepancy was observed in terms of GUS charge variants by capillary electrophoresis analysis. MALDI-TOF/TOF analysis of released N-glycans from GUS showed a minor variation in glycoforms among the treatment groups. Temperature shift resulted in a slightly increased sialylated glycan content (21.6%) when compared to control (15.5%). These results suggest that bioreactor processes have a differential effect, and better control is required for achieving improved production of GUS enzyme in CHO cells without affecting drastically its CQAs. However, the purification method allowed for enrichment of GUS with similar CQA profiles, regardless of the upstream treatments, indicating for the first time that the effect of slight alterations in upstream process parameters on the CQA profile can be offset with an effective and robust purification method downstream to maintain drug substance uniformity.


Subject(s)
Bioreactors , Biotechnology/methods , Cell Culture Techniques/methods , Glucuronidase/isolation & purification , Recombinant Proteins/isolation & purification , Animals , Butyrates/metabolism , CHO Cells , Cricetulus , Culture Media/chemistry , Female , Glucuronidase/biosynthesis , Glucuronidase/genetics , Humans , Hydrogen-Ion Concentration , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Temperature
2.
Proc Natl Acad Sci U S A ; 113(3): 704-9, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26729870

ABSTRACT

Heparanase is an endoglycosidase that cleaves heparan sulfate side chains of proteoglycans, resulting in disassembly of the extracellular matrix underlying endothelial and epithelial cells and associating with enhanced cell invasion and metastasis. Heparanase expression is induced in carcinomas and sarcomas, often associating with enhanced tumor metastasis and poor prognosis. In contrast, the function of heparanase in hematological malignancies (except myeloma) was not investigated in depth. Here, we provide evidence that heparanase is expressed by human follicular and diffused non-Hodgkin's B-lymphomas, and that heparanase inhibitors restrain the growth of tumor xenografts produced by lymphoma cell lines. Furthermore, we describe, for the first time to our knowledge, the development and characterization of heparanase-neutralizing monoclonal antibodies that inhibit cell invasion and tumor metastasis, the hallmark of heparanase activity. Using luciferase-labeled Raji lymphoma cells, we show that the heparanase-neutralizing monoclonal antibodies profoundly inhibit tumor load in the mouse bones, associating with reduced cell proliferation and angiogenesis. Notably, we found that Raji cells lack intrinsic heparanase activity, but tumor xenografts produced by this cell line exhibit typical heparanase activity, likely contributed by host cells composing the tumor microenvironment. Thus, the neutralizing monoclonal antibodies attenuate lymphoma growth by targeting heparanase in the tumor microenvironment.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Glucuronidase/immunology , Lymphoma/pathology , Animals , Antibodies, Monoclonal/pharmacology , Cell Proliferation/drug effects , Glucuronidase/isolation & purification , HEK293 Cells , Humans , Luciferases/metabolism , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Molecular Weight , Neoplasm Metastasis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Saponins/pharmacology , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
3.
Biotechnol Appl Biochem ; 65(1): 89-98, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28805269

ABSTRACT

Human heparanase (HPSE) is an enzyme that degrades the extracellular matrix. It is implicated in a multiplicity of physiological and pathological processes encouraging angiogenesis and tumor metastasis. The protein is a heterodimer composed of a subunit of 8 kDa and another of 50 kDa. The two protein subunits are noncovalently associated. The cloning and expression of the two protein subunits in Escherichia coli and their subsequent purification to homogeneity under native conditions result in the production of an active HPSE enzyme. The substrate specificity of the HPSE was studied by docking of a putative substrate that is a designed oligosaccharide with the minimum recognition backbone, with the additional 2-N-sulfate and 6-O-sulfate groups at the nonreducing GlcN and a fluorogenic tag at the reducing extremity GlcN. To develop a quantitative fluorescence assay with this substrate would be extremely useful in studies on HPSE, as the HPSE cleavage of fluorogenic tag would result in a measurable response.


Subject(s)
Cloning, Molecular , Escherichia coli/genetics , Glucuronidase/biosynthesis , Molecular Docking Simulation , Escherichia coli/metabolism , Glucuronidase/isolation & purification , Glucuronidase/metabolism , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Substrate Specificity
4.
Biotechnol Lett ; 40(1): 111-118, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28980152

ABSTRACT

OBJECTIVE: To isolate and characterize the kinetics of variants of E. coli ß-glucuronidase (GUS) having altered substrate specificity. RESULTS: Two small combinatorial libraries of E. coli GUS variants were constructed and screened for improved activities towards the substrate p-nitrophenyl-ß-D-galactoside (pNP-gal). Nine of the most active variants were purified and their kinetic parameters were determined. These variants show up to 134-fold improved kcat/KM value towards pNP-gal compared to wild-type GUS, up to 9 × 108-fold shift in specificity from p-nitrophenyl-ß-D-glucuronide (pNP-glu) to pNP-gal compared to wild-type, and 103-fold increase in specificity shift compared to a previously evolved GUS variant. CONCLUSIONS: The kinetic data collected for nine new GUS variants is invaluable for training computational protein design models that better predict amino acid substitutions which improve activity of enzyme variants having altered substrate specificity.


Subject(s)
Catalytic Domain , Escherichia coli/enzymology , Glucuronidase/genetics , Glucuronidase/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Substrate Specificity , Glucuronidase/isolation & purification , Kinetics , Mutant Proteins/isolation & purification , Nitrophenylgalactosides/metabolism
5.
Chem Biodivers ; 15(2)2018 Feb.
Article in English | MEDLINE | ID: mdl-29161468

ABSTRACT

Here, we examine the relationship between contents of principal flavones in hairy roots of Scutellaria baicalensis with the activity of the ß-glucuronidase (sGUS) enzyme during a culturing cycle. Using RP-HPLC, we show that the highest contents of aglycones, baicalin and wogonin is observed at the growth days 8, 14, and 71 and reach 45, 41, and 62% (based on the total weight of hairy roots of the Baikal skullcap), correspondingly. Their accumulation is accompanied by increase of the sGUS activity, which we determined fluorometrically. Moreover, the enzyme activity is characterized by significant and reasonable correlation only with the wogonin contents. Our results confirm a significant role of sGUS at the final steps of the metabolism in root-specific flavones of Baikal skullcap and suggest how one can optimize the conditions of culturing the hairy roots for biotechnological production of individual flavonoids. For example, at the culturing day 71 wogonin constituted over 80% of all flavones extracted from cells.


Subject(s)
Flavones/metabolism , Glucuronidase/metabolism , Plant Extracts/metabolism , Flavones/analysis , Flavones/isolation & purification , Fluorometry , Glucuronidase/isolation & purification , Molecular Structure , Plant Extracts/analysis , Plant Extracts/isolation & purification , Plant Roots/chemistry , Plant Roots/metabolism , Scutellaria baicalensis
6.
Nat Chem Biol ; 11(12): 955-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26565989

ABSTRACT

We report the structural and functional characterization of a novel heparanase (BpHep) from the invasive pathogenic bacterium Burkholderia pseudomallei (Bp), showing ∼24% sequence identity with human heparanase (hHep). Site-directed mutagenesis studies confirmed the active site resi-dues essential for activity, and we found that BpHep has specificity for heparan sulfate. Finally, we describe the first heparanase X-ray crystal structure, which provides new insight into both substrate recognition and inhibitor design.


Subject(s)
Burkholderia pseudomallei/enzymology , Glucuronidase/chemistry , Glucuronidase/metabolism , Crystallography, X-Ray , Glucuronidase/isolation & purification , Humans , Models, Molecular , Protein Conformation
7.
Protein Expr Purif ; 140: 28-35, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28734840

ABSTRACT

Human ß-glucuronidase (GUS; EC 3.2.1.31) is a lysosomal enzyme that catalyzes the hydrolysis of ß-d-glucuronic acid residues from the non-reducing termini of glycosaminoglycans. Impairment in GUS function leads to the metabolic disorder mucopolysaccharidosis type VII, also known as Sly syndrome. We produced GUS from a CHO cell line grown in suspension in a 15 L perfused bioreactor and developed a three step purification procedure that yields ∼99% pure enzyme with a recovery of more than 40%. The method can be completed in two days and has the potential to be integrated into a continuous manufacturing scheme.


Subject(s)
Glucuronidase/biosynthesis , Glucuronidase/isolation & purification , Lysosomal Storage Diseases/enzymology , Animals , CHO Cells/enzymology , Cricetulus , Glucuronidase/chemistry , Humans , Lysosomal Storage Diseases/pathology
8.
Appl Microbiol Biotechnol ; 98(9): 4021-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24253830

ABSTRACT

Baicalin (baicalein 7-O-ß-D-glucuronide) is one of the major flavonoid glucuronides found in traditional herbal medicines. Because its aglycone, baicalein, is absorbed more quickly and shows more effective properties than baicalin, the conversion of baicalin into baicalein by ß-glucuronidase (GUS) has drawn the attention of researchers. Recently, we have found that Lactobacillus brevis subsp. coagulans can convert baicalin to baicalein. Therefore, we aimed to identify and characterize the converting enzyme from L. brevis subsp. coagulans. First, we purified this enzyme from the cell-free extracts of L. brevis subsp. coagulans and cloned its gene. Surprisingly, this enzyme was found to be a GUS belonging to glycoside hydrolase (GH) family 30 (designated as LcGUS30), and its amino acid sequence has little similarity with any GUS belonging to GH families 1, 2, and 79 that have been reported so far. We then established a high-level expression and simple purification system of the recombinant LcGUS30 in Escherichia coli. The detailed analysis of the substrate specificity revealed that LcGUS30 has strict specificity toward glycon but not toward aglycones. Interestingly, LcGUS30 prefers baicalin rather than estrone 3-(ß-D-glucuronide), one of the human endogenous steroid hormones. These results indicated that L. brevis subsp. coagulans and LcGUS30 should serve as powerful tools for the construction of a safe bioconversion system for baicalin. In addition, we propose that this novel type of GUS forms a new group in subfamily 3 of GH family 30.


Subject(s)
Flavanones/metabolism , Flavonoids/metabolism , Glucuronidase/isolation & purification , Glucuronidase/metabolism , Glycoside Hydrolases/isolation & purification , Glycoside Hydrolases/metabolism , Levilactobacillus brevis/enzymology , Amino Acid Sequence , Biotransformation , Cloning, Molecular , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/genetics , Estrone/analogs & derivatives , Estrone/metabolism , Gene Expression , Glucuronidase/genetics , Glycoside Hydrolases/genetics , Hydrolysis , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Substrate Specificity
9.
Biomed Chromatogr ; 27(10): 1280-95, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23649485

ABSTRACT

Glucuronidation and sulfation represent two major pathways in phase II drug metabolism in humans and other mammalian species. The great majority of drugs, for example, polyphenols, flavonoids and anthraquinones, could be transformed into sulfated and glucuronidated conjugates simultaneously and extensively in vivo. The pharmacological activities of drug conjugations are normally decreased compared with those of their free forms. However, some drug conjugates may either bear biological activities themselves or serve as excellent sources of biologically active compounds. As the bioactivities of drugs are thought to be relevant to the kinetics of their conjugates, it is essential to study the pharmacokinetic behaviors of the conjugates in more detail. Unfortunately, the free forms of drugs cannot be detected directly in most cases if their glucuronides and sulfates are the predominant forms in biological samples. Nevertheless, an initial enzymatic hydrolysis step using ß-glucuronidase and/or sulfatase is usually performed to convert the glucuronidated and/or sulfated conjugates to their free forms prior to the extraction, purification and other subsequent analysis steps in the literature. This review provides fundamental information on drug metabolism pathways, the bio-analytical strategies for the quantification of various drug conjugates, and the applications of the analytical methods to pharmacokinetic studies.


Subject(s)
Glucuronidase/analysis , Pharmaceutical Preparations/analysis , Pharmaceutical Preparations/metabolism , Sulfatases/analysis , Animals , Chemical Fractionation , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Glucuronidase/chemistry , Glucuronidase/isolation & purification , Glucuronidase/metabolism , Humans , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/isolation & purification , Sulfatases/chemistry , Sulfatases/isolation & purification , Sulfatases/metabolism
10.
Bioconjug Chem ; 22(5): 938-48, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21443266

ABSTRACT

Antibody-directed enzyme prodrug therapy (ADEPT) utilizing ß-glucuronidase is a promising method to enhance the therapeutic index of cancer chemotherapy. In this approach, an immunoenzyme (antibody-ß-glucuronidase fusion protein) is employed to selectively activate anticancer glucuronide prodrugs in the tumor microenvironment. A major roadblock to the clinical translation of this therapeutic strategy, however, is the low enzymatic activity and strong immunogenicity of the current generation of immunoenzymes. To overcome this problem, we fused a humanized single-chain antibody (scFv) of mAb CC49 to S2, a human ß-glucuronidase (hßG) variant that displays enhanced catalytic activity for prodrug hydrolysis. Here, we show that hcc49-S2 displayed 100-fold greater binding avidity than hcc49 scFv, possessed greater enzymatic activity than wild-type hßG, and more effectively killed antigen-positive cancer cells exposed to an anticancer glucuronide prodrug as compared to an analogous hßG immunoenzyme. Treatment of tumor-bearing mice with hcc49-S2 followed by prodrug significantly delayed tumor growth as compared to hcc49-hßG. Our study shows that hcc49-S2 is a promising targeted enzyme for cancer treatment and demonstrates that enhancement of human enzyme catalytic activity is a powerful approach to improve immunoenzyme efficacy.


Subject(s)
Antibodies, Neoplasm/metabolism , Glucuronidase/metabolism , Glucuronides/metabolism , Prodrugs/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Microenvironment , Animals , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/isolation & purification , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biocatalysis , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Glucuronidase/chemistry , Glucuronidase/isolation & purification , Glucuronides/pharmacology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Molecular Imaging , NIH 3T3 Cells , Prodrugs/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Structure-Activity Relationship , Xenograft Model Antitumor Assays
11.
Proc Natl Acad Sci U S A ; 105(7): 2616-21, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18268347

ABSTRACT

Enzyme replacement therapy has been used successfully in many lysosomal storage diseases. However, correction of brain storage has been limited by the inability of infused enzyme to cross the blood-brain barrier. The newborn mouse is an exception because recombinant enzyme is delivered to neonatal brain after mannose 6-phosphate receptor-mediated transcytosis. Access to this route is very limited after 2 weeks of age. Recently, several studies showed that multiple infusions of high doses of enzyme partially cleared storage in adult brain. These results raised the question of whether correction of brain storage by repeated high doses of enzyme depends on mannose 6-phosphate receptor-mediated uptake or whether enzyme gains access to brain storage by another route when brain capillaries are exposed to prolonged, high levels of circulating enzyme. To address this question, we used an enzyme whose carbohydrate-dependent receptor-mediated uptake was inactivated by chemical modification. Treatment of human beta-glucuronidase (GUS) with sodium metaperiodate followed by sodium borohydride reduction (PerT-GUS) eliminated uptake by mannose 6-phosphate and mannose receptors in cultured cells and dramatically slowed its plasma clearance from a t(1/2) of <10 min to 18 h. Surprisingly, PerT-GUS infused weekly for 12 weeks was more effective in clearing central nervous system storage than native GUS at the same dose. In fact, PerT-GUS resulted in almost complete reversal of storage in neocortical and hippocampal neurons. This enhanced correction of neuronal storage by long-circulating enzyme, which targets no known receptor, suggests a delivery system across the blood-brain barrier that might be exploited therapeutically.


Subject(s)
Blood-Brain Barrier/drug effects , Glucuronidase/chemistry , Glucuronidase/therapeutic use , Mucopolysaccharidosis VII/drug therapy , Neurons/drug effects , Neurons/enzymology , beta-Glucosidase/chemistry , beta-Glucosidase/therapeutic use , Animals , Borohydrides/therapeutic use , Cells, Cultured , Enzyme Stability , Glucuronidase/genetics , Glucuronidase/isolation & purification , Humans , Mice , Mucopolysaccharidosis VII/enzymology , Mucopolysaccharidosis VII/pathology , Periodic Acid/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Temperature , beta-Glucosidase/genetics , beta-Glucosidase/isolation & purification
12.
Int J Biol Macromol ; 159: 782-792, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32416297

ABSTRACT

Glycyrrhetinic acid monoglucuronide (GAMG) is an innovative functional sweetener with higher sweetness and stronger pharmacological activity than glycyrrhizin (GL). A novel ß-glucuronidase (cg-GUS) was firstly screened from plant endophytic fungus Chaetomium globosum DX-THS3. The cg-GUS demonstrated the specify and highly transform glycyrrhizin (GL) to generate GAMG, and the maximum activity of ß-glucuronidase at 45 °C and pH 6.0, displaying excellent thermostability and pH-stability. The Km and Vmax values of cg-GUS were 0.134 mM and 236.42 mM/min/mg, respectively, which showed the high chemical bond selectivity and biotransformation efficiency of cg-GUS. Meanwhile, the cg-GUS gene (1896 bp) was analyzed, and Gly-345, Ser-539, Gly-563, Ala-579, Ser-581 and Glu-619 in GH2 catalytic domain of cg-GUS are potential mutation position for result in high-efficient and substrate-specify of cg-GUS. Our results were indicated that cg-GUS is a biocatalyst for production of GAMG and potent application in food and medicinal industry.


Subject(s)
Chaetomium/enzymology , Glucuronidase/chemistry , Glucuronidase/isolation & purification , Amino Acid Sequence , Catalysis , Chaetomium/genetics , Chromatography, High Pressure Liquid , Cloning, Molecular , Enzyme Activation , Gene Expression , Glucuronidase/genetics , Glucuronides/metabolism , Glycyrrhizic Acid/metabolism , Kinetics , Recombinant Proteins
13.
J Cell Biol ; 100(6): 1894-904, 1985 Jun.
Article in English | MEDLINE | ID: mdl-3922995

ABSTRACT

Lysosomal enzymes have been shown to be synthesized as microsomal precursors, which are processed to mature enzymes located in lysosomes. We examined the effect of ammonium chloride on the intracellular processing and secretion of two lysosomal enzymes, beta-glucuronidase and beta-galactosidase, in mouse macrophages. This lysosomotropic drug caused extensive secretion of both precursor and mature enzyme forms within a few hours, as documented by pulse radiolabeling and molecular weight analysis. The normal intracellular route for processing and secretion of precursor enzyme was altered in treated cells. A small percentage of each precursor was delivered to the lysosomal organelle slowly. Most precursor forms traversed the Golgi apparatus, underwent further processing of carbohydrate moieties, and were then secreted in a manner similar to secretory proteins. The lag time for secretion of newly synthesized beta-galactosidase precursor was notably longer than that for the beta-glucuronidase precursor. The source of the secreted mature enzyme was the lysosomal organelle. Macrophages from the pale ear mutant were markedly deficient in secretion of mature lysosomal enzyme but secreted precursor forms normally. These results suggest that ammonia-treated macrophages contain two distinct intracellular pathways for secretion of lysosomal enzymes and that a specific block in the release of lysosomal contents occurs in the pale ear mutant.


Subject(s)
Ammonium Chloride/pharmacology , Galactosidases/metabolism , Glucuronidase/metabolism , Lysosomes/enzymology , Macrophages/enzymology , beta-Galactosidase/metabolism , Animals , Female , Glucuronidase/genetics , Glucuronidase/isolation & purification , Kinetics , Lysosomes/drug effects , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Weight , Mutation , Protein Precursors/metabolism , Subcellular Fractions/enzymology , beta-Galactosidase/genetics , beta-Galactosidase/isolation & purification
14.
J Anal Toxicol ; 43(3): 221-227, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30517702

ABSTRACT

Pain management laboratories analyze biological fluids (urine, saliva or blood) from patients treated for chronic pain to ensure compliance and to detect undisclosed drug use. The quantitation of multi-panel drugs in urine and tissues utilizes ß-glucuronidase to cleave the glucuronic acid and liberate the parent drug for mass spectrometry analysis. This work focuses on the comparison of three different, purified and commercially available ß-glucuronidases across 83 patient urine samples. One enzyme is genetically modified, expressed in bacteria and the other two enzymes are purified from abalone. The results indicate that the source of ß-glucuronidase plays an important role in substrate specificity which in turn dictates hydrolysis efficiency. Contaminants in the enzyme solutions also interfere with analyte detection. Altogether, these factors impact precision and accuracy of data interpretation, leading up to 13% positive/negative disagreement.


Subject(s)
Analgesics, Opioid/urine , Glucuronidase/isolation & purification , Glucuronidase/metabolism , Glucuronides/metabolism , Illicit Drugs/urine , Substance Abuse Detection/methods , Analgesics, Opioid/metabolism , Calibration , Chromatography, High Pressure Liquid , Humans , Hydrolysis , Illicit Drugs/metabolism , Patient Compliance , Reference Standards , Reproducibility of Results , Substance Abuse Detection/instrumentation , Tandem Mass Spectrometry
15.
Int J Biochem Cell Biol ; 40(3): 530-42, 2008.
Article in English | MEDLINE | ID: mdl-17981072

ABSTRACT

Heparanase is an endoglycosidase which cleaves heparan sulfate and hence participates in degradation and remodeling of the extracellular matrix. Importantly, heparanase activity correlated with the metastatic potential of tumor-derived cells, attributed to enhanced cell dissemination as a consequence of heparan sulfate cleavage and remodeling of the extracellular matrix barrier. Heparanase has been characterized as a glycoprotein, yet glycan biochemical analysis was not performed to date. Here, we applied the Qproteometrade mark GlycoArray kit to perform glycan analysis of heparanase, and compared the kit results with the more commonly used biochemical analyses. We employed fibroblasts isolated from patients with I-cell disease (mucolipidosis II), fibroblasts deficient of low density lipoprotein receptor-related protein and fibroblasts lacking mannose 6-phosphate receptor, to explore the role of mannose 6-phosphate in heparanase uptake. Iodinated heparanase has been utilized to calculate binding affinity. We provide evidence for hierarchy of binding to cellular receptors as a function of heparanase concentration. We report the existence of a high affinity, low abundant (i.e., low density lipoprotein receptor-related protein, mannose 6-phosphate receptor), as well as a low affinity, high abundant (i.e., heparan sulfate proteoglycan) receptors that mediate heparanase binding, and suggest that these receptors co-operate to establish high affinity binding sites for heparanase, thus maintaining extracellular retention of the enzyme tightly regulated.


Subject(s)
Fibroblasts/metabolism , Glucuronidase/metabolism , Heparitin Sulfate/metabolism , Mannosephosphates/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Line, Tumor , Cricetinae , Extracellular Matrix/enzymology , Extracellular Matrix/metabolism , Fibroblasts/enzymology , Glucuronidase/isolation & purification , Humans , Mucolipidoses/enzymology , Mucolipidoses/metabolism , Recombinant Proteins
16.
Plant Cell Physiol ; 49(9): 1331-41, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18667448

ABSTRACT

Beta-glucuronidase (GUS) activities have been extensively characterized in bacteria, fungi, and animals, and the bacterial enzyme GUSA from Escherichia coli is commonly used as a reporter for gene expression studies in plants. Although endogenous GUS activity has been observed in plants, the nature and function of the enzymes involved remain elusive. Here we report on tissue-specific localization, partial purification and identification of AtGUS2, a GUS active under acidic conditions from Arabidopsis thaliana. This enzyme belongs to the GH79 family in the Carbohydrate-Active Enzymes database, which also includes mammalian heparanases that degrade the carbohydrate moieties of cell surface proteoglycans, and fungal enzymes active on arabinogalactan proteins (AGPs). We characterized a knockout insertion line (atgus2-1) and transgenic lines overexpressing AtGUS2 (Pro(35S):AtGUS2). Endogenous GUS activity assayed histochemically and biochemically was absent in atgus2-1 tissues and four times higher in Pro(35S):AtGUS2 lines. AGPs purified from atgus2-1 and Pro(35S):AtGUS2 seedlings showed higher and markedly lower glucuronic acid content, respectively. Our results suggest that endogenous GUS activity influences the sugar composition of the complex polysaccharide chains of AGPs. We also show that transgenics display hypocotyl and root growth defects compared to wild-type plants. Hypocotyl and root lengths are increased in Pro(35S):AtGUS2 seedlings, whereas hypocoyl length is reduced in atgus2-1 seedlings. These data are consistent with a role for the carbohydrate moieties of AGPs in cell growth.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Arabidopsis/genetics , Glucuronidase/metabolism , Amino Acid Sequence , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Chromatography , Cloning, Molecular , Genes, Plant , Glucuronic Acid/metabolism , Glucuronidase/genetics , Glucuronidase/isolation & purification , Hydrogen-Ion Concentration , Hypocotyl/growth & development , Molecular Sequence Data , Mucoproteins/isolation & purification , Mucoproteins/metabolism , Plant Proteins/isolation & purification , Plant Proteins/metabolism , Plant Roots/growth & development , Plants, Genetically Modified/enzymology , Plants, Genetically Modified/genetics , Polysaccharides/metabolism , RNA, Plant/genetics , Temperature , Transformation, Genetic
17.
J Immunol Methods ; 331(1-2): 82-93, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18162185

ABSTRACT

Degradation of extracellular matrix is associated with extravasation of metastatic tumor cells and inflammatory cells. Heparanase, the heparan sulfate-specific endo-beta-glucuronidase, is a key enzyme for the matrix degradation, yet its involvement in extravasation and invasion during pathological processes was not fully clarified in vivo. In the present study, we examined heparanase expression in mouse experimental models, lung metastasis of melanoma and skin infiltration of neutrophils. Sixteen novel monoclonal antibodies specific for mouse heparanase were established by enzyme-linked immunosorbent assay with a recombinant mouse proheparanase, immunocytochemical staining of B16F10 melanoma cells cultured in vitro, and immunoprecipitation of the lysate of heparanase transfectant cells. Heparanase expression in metastatic nodules of B16F10 melanoma cells and in neutrophils localized in the inflamed skin was immunohistochemically detected using a monoclonal antibody RIO-1 that recognized the C-terminus of mouse heparanase. Homogeneous and strong heparanase staining was observed in 46% of the lung micrometastases of B16F10 melanoma cells. The staining was intensely positive on the invasive front of larger established metastasis nodules, but it was weak or heterogeneous inside the nodules. Heparanase expression in skin-infiltrating neutrophils was examined after inducing local inflammation with croton oil. The monoclonal antibody stained a significant portion of neutrophils inside and along the blood vessels, whereas it did not stain dermal neutrophils located distant from the vasculatures. The present study strongly suggests that both melanoma cells and neutrophils transiently express heparanase before and during the invasive process in vivo.


Subject(s)
Antibodies, Monoclonal/immunology , Extracellular Matrix/metabolism , Glucuronidase/metabolism , Immunoassay/methods , Melanoma, Experimental/enzymology , Neutrophils/enzymology , Animals , Dermatitis/enzymology , Female , Glucuronidase/immunology , Glucuronidase/isolation & purification , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neutrophils/cytology , Neutrophils/metabolism , Rats , Rats, Wistar , Recombinant Proteins
18.
Biotechnol Bioeng ; 99(4): 902-9, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-17787005

ABSTRACT

Tobacco has proven to be a promising alternative for the production of recombinant therapeutic proteins and offers numerous advantages over other plants as a host system. However, the recovery and purification steps needed to obtain a protein at high recovery and purity have not been well investigated. In this study, a process was developed to purify a model acidic protein, recombinant beta-glucuronidase (rGUS) from transgenic tobacco leaf tissue, in three main steps after extraction: polyelectrolyte precipitation, hydrophobic interaction chromatography (HIC), and hydroxyapatite chromatography (HAC). Using this three-step process, up to 40% of the initial rGUS activity could be recovered to near homogeneity as judged by SDS-PAGE. This work demonstrates that acidic recombinant proteins expressed in tobacco may be purified to high yield with high purity in a minimal amount of steps that are suitable for scale-up. Furthermore, the general steps used in this process may suggest that a wide variety of acidic recombinant proteins may be purified in a similar manner from transgenic tobacco or other leafy crops.


Subject(s)
Glucuronidase/isolation & purification , Glucuronidase/physiology , Nicotiana/physiology , Plant Leaves/metabolism , Plants, Genetically Modified/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Chromatography/methods
19.
J Chromatogr A ; 1142(1): 98-105, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-16950325

ABSTRACT

Tobacco has been studied as a possible host for the production of recombinant proteins. In this report, recombinant beta-glucuronidase (rGUS) was used as a model protein to study the feasibility of using polyethyleneimine (PEI) precipitation to fractionate acidic recombinant proteins from transgenic tobacco. Results showed that rGUS was preferentially precipitated when the PEI dosage was beyond 200mg PEI/g total protein. At 700-800 mg PEI/g total protein, nearly 100% rGUS was precipitated with less than 40% native tobacco proteins co-precipitated. Approximately 85-90% of the rGUS activity could be recovered from the precipitation pellet for an enrichment ratio of 2.7-3.4. As a comparison, anion exchange chromatography (AEX) yielded similar results to PEI precipitation with 66-90% rGUS activity recovered and an enrichment ratio of 1.8-3.1. The rGUS was further purified by an additional hydrophobic interaction chromatographic (HIC) step after precipitation or AEX. Similar results in terms of rGUS activity recovered and enrichment were obtained. The major interfering protein at the end of all purification steps is most likely the Fraction 1 protein ribulose 1,5-bisphosphate carboxylase-oxygenase (Rubisco). The presence of this protein is likely the cause for the varying and somewhat low enrichment ratios, but it may be later removed via a size-exclusion chromatography step. PEI precipitation offers the advantage of allowing significant sample concentration prior to subsequent purification techniques such as chromatography and is much less expensive than chromatographic methods as well. Through direct comparison, this study shows that PEI may be used as an initial fractionation step in replacement of AEX to facilitate the purification of acidic recombinant proteins from transgenic tobacco.


Subject(s)
Chromatography, Ion Exchange/methods , Glucuronidase/isolation & purification , Nicotiana/enzymology , Plants, Genetically Modified/enzymology , Animals , Chemical Precipitation , Recombinant Proteins/isolation & purification , Nicotiana/genetics
20.
Protein Pept Lett ; 14(10): 1008-13, 2007.
Article in English | MEDLINE | ID: mdl-18220999

ABSTRACT

This paper studies the beta-glucuronidase in the mollusk Pomacea sp. The beta-glucuronidase was isolated 206-fold with a 1,5% yield and the cinetc parameters was: pH 5.0, 65 degrees C, K(m) of 72 x 10(-2) mM and molecular mass of 116 kDa. HPLC confirmed the purity. BaCl(2) increased beta-glucuronidase activity and SDS and NaH(2)PO(4) inhibited completely.


Subject(s)
Gastropoda/enzymology , Glucuronidase/isolation & purification , Glucuronidase/metabolism , Animals , Cattle , Chondroitin Sulfates/metabolism , Glucuronidase/chemistry , Hydrolysis/drug effects , Kinetics , Solubility , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL