Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Cancer ; 5(6): 895-915, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38448522

ABSTRACT

Gemcitabine is a potent inhibitor of DNA replication and is a mainstay therapeutic for diverse cancers, particularly pancreatic ductal adenocarcinoma (PDAC). However, most tumors remain refractory to gemcitabine therapies. Here, to define the cancer cell response to gemcitabine, we performed genome-scale CRISPR-Cas9 chemical-genetic screens in PDAC cells and found selective loss of cell fitness upon disruption of the cytidine deaminases APOBEC3C and APOBEC3D. Following gemcitabine treatment, APOBEC3C and APOBEC3D promote DNA replication stress resistance and cell survival by deaminating cytidines in the nuclear genome to ensure DNA replication fork restart and repair in PDAC cells. We provide evidence that the chemical-genetic interaction between APOBEC3C or APOBEC3D and gemcitabine is absent in nontransformed cells but is recapitulated across different PDAC cell lines, in PDAC organoids and in PDAC xenografts. Thus, we uncover roles for APOBEC3C and APOBEC3D in DNA replication stress resistance and offer plausible targets for improving gemcitabine-based therapies for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Cytidine Deaminase , DNA Replication , Deoxycytidine , Gemcitabine , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Cell Line, Tumor , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Mice , Drug Resistance, Neoplasm/genetics , Antimetabolites, Antineoplastic/pharmacology , Xenograft Model Antitumor Assays , CRISPR-Cas Systems
2.
Oncotarget ; 8(2): 3064-3071, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27902970

ABSTRACT

BACKGROUND: Polo-like kinase 4 (PLK4) plays a key role in centriole replication. Hence PLK4 inhibition disrupts mitosis, and offers a novel approach to treating chromosomally unstable cancers, including pancreatic cancer. CFI-400945 is a first in class small molecule PLK4 inhibitor, currently undergoing early phase clinical trials. RESULTS: Treatment with CFI-400945 significantly reduced tumor growth and increased survival in four out of the six models tested. Consistent with PLK4 inhibition, we observed reduced expression of the proliferation marker Ki-67 associated with an increase in nuclear diameter during treatment with CFI-400945. Additionally, treatment with CFI-400945 resulted in a significant reduction of tumor-initiating cells. DISCUSSION: These results support the further investigation of PLK4 as a drug target in pancreatic cancer. METHODS: Sensitivity to CFI-400945 was tested in a series of six patient-derived pancreatic cancer xenografts, selected to represent the range of growth characteristics, genetic features, and hypoxia found in pancreatic cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Indazoles/pharmacology , Indoles/pharmacology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Humans , Immunohistochemistry , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Burden , Xenograft Model Antitumor Assays
3.
PLoS One ; 11(12): e0167272, 2016.
Article in English | MEDLINE | ID: mdl-28033382

ABSTRACT

BACKGROUND: The BRCA2 gene product plays an important role in DNA double strand break repair. Therefore, we asked whether radiation sensitivity of pancreatic cancers developing in individuals with germline BRCA2 mutations can be enhanced by agents that inhibit poly (ADP-ribose) polymerase (PARP). METHODS: We compared the sensitivity of two patient-derived pancreatic cancer xenografts, expressing a truncated or wild type BRCA 2, to ionizing radiation alone or in combination with olaparib (AZD-2281). Animals were treated with either a single dose of 12Gy, 7 days of olaparib or 7 days of olaparib followed by a single dose of 12Gy. Response was assessed by tumour growth delay and the activation of damage response pathways. RESULTS: The BRCA2 mutated and wild type tumours showed similar radiation sensitivity, and treatment with olaparib did not further sensitize either model when compared to IR alone. CONCLUSIONS: While PARP inhibition has been shown to be effective in BRCA-mutated breast and ovarian cancers, it is less well established in pancreatic cancer patients. Our results show no radiosensitization in a germline BRCA 2 mutant and suggest that combining PARP inhibition and IR may not be beneficial in BRCA 2 related pancreatic tumors.


Subject(s)
BRCA2 Protein/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy/methods , Humans , Mice , Mice, SCID , Pancreatic Neoplasms/genetics , Radiation, Ionizing , Radiation-Sensitizing Agents/therapeutic use
4.
Oncotarget ; 7(23): 33571-80, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27248663

ABSTRACT

Previous reports have suggested that the hypoxic microenvironment provides a niche that supports tumor stem cells, and that this might explain clinical observations linking hypoxia to metastasis. To test this, we examined the effects of a hypoxia-activated prodrug, TH-302, on the tumor-initiating cell (TIC) frequency of patient-derived pancreatic xenografts (PDX).The frequencies of TIC, measured by limiting dilution assay, varied widely in 11 PDX models, and were correlated with rapid growth but not with the levels of hypoxia. Treatment with either TH-302 or ionizing radiation (IR), to target hypoxic and well-oxygenated regions, respectively, reduced TIC frequency, and the combination of TH-302 and IR was much more effective in all models tested. The combination was also more effective than TH-302 or IR alone controlling tumor growth, particularly treating the more rapidly-growing/hypoxic models. These findings support the clinical utility of hypoxia targeting in combination with radiotherapy to treat pancreatic cancers, but do not provide strong evidence for a hypoxic stem cell niche.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Hypoxia/drug effects , Nitroimidazoles/pharmacology , Pancreatic Neoplasms/pathology , Phosphoramide Mustards/pharmacology , Tumor Microenvironment/drug effects , Animals , Cell Line, Tumor , Humans , Mice , Neoplastic Stem Cells/drug effects , Prodrugs/pharmacology , Xenograft Model Antitumor Assays
5.
Mol Cancer Ther ; 3(10): 1239-48, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15486191

ABSTRACT

Anticancer effects of the dietary isothiocyanate sulforaphane were investigated in the human pancreatic cancer cell lines MIA PaCa-2 and PANC-1. Sulforaphane-treated cells accumulated in metaphase as determined by flow cytometry [4C DNA content, cyclin A(-), cyclin B1(+), and phospho-histone H3 (Ser(10))(+)]. In addition, treated cells showed nuclear apoptotic morphology that coincided with an activation of caspase-8, loss of mitochondrial membrane potential, and loss of plasma membrane integrity. The initial detection of caspase-3 cleavage occurring in G(2)-M arrest was independent of a change in phospho-cdc2 (Tyr(15)) protein; consequently, sulforaphane treatment combined with UCN-01 had no significant impact on cellular toxicity. Incubations at higher sulforaphane doses (>10 micromol/L) resulted in cleavage of caspase-3 in the G(1) subpopulation, suggesting that the induction of apoptosis and the sulforaphane-induced mitosis delay at the lower dose are independently regulated. Cellular toxicity in MIA PaCa-2, and to a greater extent in PANC-1, was positively correlated with a decrease in cellular glutathione levels, whereas sustained increases in glutathione observed in MIA PaCa-2 cells or the simultaneous incubation with N-acetyl-L-cysteine in PANC-1 cells were associated with resistance to sulforaphane-induced apoptosis. Daily sulforaphane i.p. injections (375 micromol/kg/d for 3 weeks) in severe combined immunodeficient mice with PANC-1 s.c. tumors resulted in a decrease of mean tumor volume by 40% compared with vehicle-treated controls. Our findings suggest that, in addition to the known effects on cancer prevention, sulforaphane may have activity in established pancreatic cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis , Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Staurosporine/analogs & derivatives , Thiocyanates/pharmacology , Animals , Antigens/chemistry , Blotting, Western , Caspase 3 , Caspase 8 , Caspases/metabolism , Cell Cycle , Cell Line, Tumor , Cell Membrane/metabolism , Cell Nucleus/metabolism , DNA/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Flow Cytometry , Glutathione/metabolism , Humans , Isothiocyanates , Male , Membrane Potentials , Mice , Mice, SCID , Microscopy, Fluorescence , Mitochondria/metabolism , Mitosis , Neoplasm Transplantation , Oxidative Stress , Reactive Oxygen Species , Staurosporine/pharmacology , Sulfoxides , Time Factors , Tyrosine/chemistry
6.
J Control Release ; 206: 122-30, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25804872

ABSTRACT

Pancreatic ductal adenocarcinomas are characterized by the desmoplastic reaction, a dense fibrous stroma that has been shown to be supportive of tumor cell growth, invasion, and metastasis, and has been associated with resistance to chemotherapy and reduced patient survival. Here, we investigated targeted depletion of stroma for pancreatic cancer therapy via taxane nanoparticles. Cellax-DTX polymer is a conjugate of docetaxel (DTX), polyethylene glycol (PEG), and acetylated carboxymethylcellulose, a construct which condenses into well-defined 120nm particles in an aqueous solution, and is suitable for intravenous injection. We examined Cellax-DTX treatment effects in highly stromal primary patient-derived pancreatic cancer xenografts and in a metastatic PAN02 mouse model of pancreatic cancer, focusing on specific cellular interactions in the stroma, pancreatic tumor growth and metastasis. Greater than 90% of Cellax-DTX particles accumulate in smooth muscle actin (SMA) positive cancer-associated fibroblasts which results in long-term depletion of this stromal cell population, an effect not observed with Nab-paclitaxel (Nab-PTX). The reduction in stromal density leads to a >10-fold increase in tumor perfusion, reduced tumor weight and a reduction in metastasis. Consentingly, Cellax-DTX treatment increased survival when compared to treatment with gemcitabine or Nab-PTX in a metastatic PAN02 mouse model. Cellax-DTX nanoparticles interact with the tumor-associated stroma, selectively interacting with and depleting SMA positive cells and macrophage, effects of which are associated with significant changes in tumor progression and metastasis.


Subject(s)
Antineoplastic Agents/administration & dosage , Carboxymethylcellulose Sodium/chemistry , Fibroblasts/drug effects , Nanoparticles/economics , Pancreas/drug effects , Pancreatic Neoplasms/drug therapy , Taxoids/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Docetaxel , Drug Delivery Systems , Female , Fibroblasts/pathology , Humans , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Pancreas/pathology , Pancreatic Neoplasms/pathology , Polyethylene Glycols/chemistry , Taxoids/chemistry , Taxoids/therapeutic use , Xenograft Model Antitumor Assays
7.
Clin Biochem ; 36(5): 339-44, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12849864

ABSTRACT

OBJECTIVES: Mutations disrupting the interaction of extra-cellular ligands and alpha-dystroglycan are responsible for an etiologically heterogeneous group of autosomal recessive congenital muscular dystrophies (CMD) that can have associated brain and eye abnormalities. The objective is to develop a diagnostic test for one of these CMDs, Muscle-Eye-Brain disease (MEB), due to mutations in the gene encoding Protein O-Mannosyl beta-1,2-N-acetylglucosaminyltransferase 1 (POMGnT1). DESIGN AND METHODS: POMGnT1 enzyme activity was determined in extracts of muscle biopsies from four MEB patients and various controls using commercially available reagents. RESULTS: All four MEB muscle samples showed a highly significant decrease in POMGnT1 activity relative to controls. CONCLUSIONS: The assay of POMGnT1 activity in MEB muscle provides a rapid and relatively simple diagnostic test for this disease. CMDs associated with brain malformations such as MEB, WWS and FCMD are heterogenous in clinical presentation and on radiologic examination, suggesting that POMGnT1 assays of muscle biopsies should be used as a screening procedure for MEB in all CMD patients associated with brain malformations.


Subject(s)
Brain/abnormalities , Eye Abnormalities , Muscle, Skeletal/abnormalities , Muscular Dystrophies/diagnosis , N-Acetylglucosaminyltransferases/deficiency , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/metabolism , Dystroglycans , Glucuronosyltransferase/deficiency , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Ligands , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Muscular Dystrophies/congenital , Muscular Dystrophies/enzymology , Mutation , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism
8.
Zhonghua Zhong Liu Za Zhi ; 24(5): 508-10, 2002 Sep.
Article in Zh | MEDLINE | ID: mdl-12485512

ABSTRACT

OBJECTIVE: To evaluate the feasibility of surgical management for patients with locally advanced carcinomas of uterine cervix after radical radiation therapy who were prone to develop central recurrence. METHODS: These 40 patients were treated by combined pre-operative radiotherapy with dose at point A of > 70 Gy in 30 patients, 60 approximately 70 Gy in 7, 50 approximately 59 Gy in 2 and 44 Gy in 1. The interval between radiation and surgery was 1 - 6 weeks. Extrafascial hysterectomy was performed in 15 patients, subradical hysterectomy in 23 and radical hysterectomy with pelvic lymphadenectomy in 2 cases. RESULTS: These patients have been followed up for 1 - 8 years with 2 died of other diseases and 12 died of cancer. Eighteen of the 26 survivors have been followed up for more than 5 years. The 3- and 5-year survival rates were 74.9% and 66.8%. Half of the death occurred within the first year after treatment. The 2-year death rate was 9/12 (75.0%). Three patients suffered from long term complications after the treatment, but all were cured by conservative management. CONCLUSION: The combination of hysterectomy performed shortly after radical radiotherapy, ie, for patients with locally poor prognostic cervical carcinoma is reasonable and feasible.


Subject(s)
Uterine Cervical Neoplasms/radiotherapy , Uterine Cervical Neoplasms/surgery , Adult , Cervix Uteri , Combined Modality Therapy , Female , Humans , Hysterectomy , Middle Aged , Neoplasm Staging , Survival Rate , Uterine Cervical Neoplasms/mortality
9.
Clin Cancer Res ; 19(1): 118-27, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23091117

ABSTRACT

PURPOSE: MEK inhibition has clinical activity against biliary cancers and might therefore be successfully combined with gemcitabine, one of the most active chemotherapy agents for these cancers. As gemcitabine is active in S-phase, and the extracellular signal-regulated kinase (ERK) pathway has a major role driving cell-cycle progression, concurrent use of a MEK inhibitor could potentially antagonize the effect of gemcitabine. We therefore tested the sequence dependence of the combination of gemcitabine and the MEK inhibitor AZD6244 using a series of biliary cancer models. EXPERIMENTAL DESIGN: Primary xenografts were established from patients with gallbladder and distal bile duct cancer and grown in severe combined immunodeficient (SCID) mice at the subcutaneous site. Plasma and tumor drug levels and the time course for recovery of ERK signaling and S-phase were measured in tumor-bearing mice treated for 48 hours with AZD6244 and then monitored for 48 hours off treatment. On the basis of these results, two different treatment schedules combining AZD6244 with gemcitabine were tested in four different biliary cancer models. RESULTS: DNA synthesis was suppressed during treatment with AZD6244, and reentry into S-phase was delayed by approximately 48 hours after treatment. Strong schedule dependence was seen in all four biliary cancer models tested, suggesting that combined treatment with AZD6244 plus gemcitabine would be more active in patients with biliary cancer when gemcitabine is given following a 48-hour interruption in AZD6244 dosing, rather than concurrently. CONCLUSIONS: The combination of AZD6244 plus gemcitabine is highly schedule dependent, and predicted to be more effective in the clinic using sequential rather than simultaneous dosing protocols.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Biliary Tract Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Biliary Tract Neoplasms/enzymology , Biliary Tract Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice , Mice, SCID , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
10.
PLoS One ; 7(7): e41102, 2012.
Article in English | MEDLINE | ID: mdl-22829912

ABSTRACT

Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) induces apoptosis in a variety of cancer cell lines with little or no effect on normal cells. However, its effect is limited as some cancers including pancreatic cancer show de novo resistance to TRAIL induced apoptosis. In this study we report that GSK-3 inhibition using the pharmacologic agent AR-18, enhanced TRAIL sensitivity in a range of pancreatic and prostate cancer cell lines. This sensitization was found to be caspase-dependent, and both pharmacological and genetic knock-down of GSK-3 isoforms resulted in apoptotic features as shown by cleavage of PARP and caspase-3. Elevated levels of reactive oxygen intermediates and disturbance of mitochondrial membrane potential point to a mitochondrial amplification loop for TRAIL-induced apoptosis after GSK-3 inhibition. Consistent with this, overexpression of anti-apoptotic mitochondrial targets such as Bcl-XL, Mcl-1, and Bcl-2 rescued PANC-1 and PPC-1 cells from TRAIL sensitization. However, overexpression of the caspase-8 inhibitor CrmA also inhibited the sensitizing effects of GSK-3 inhibitor, suggesting an additional role for GSK-3 that inhibits death receptor signaling. Acute treatment of mice bearing PANC-1 xenografts with a combination of AR-18 and TRAIL also resulted in a significant increase in apoptosis, as measured by caspase-3 cleavage. Sensitization to TRAIL occurred despite an increase in ß-catenin due to GSK-3 inhibition, suggesting that the approach might be effective even in cancers with dysregulated ß-catenin. These results suggest that GSK-3 inhibitors might be effectively combined with TRAIL for the treatment of pancreatic cancer.


Subject(s)
Apoptosis/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Pancreatic Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/genetics , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
11.
Pancreas ; 35(3): e16-26, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17895832

ABSTRACT

OBJECTIVE: To understand the impact of protein kinase B (PKB; Akt) signaling on growth and protection from apoptosis in pancreatic ductal adenocarcinoma models demonstrating differences in PKB activity. METHODS: Gemcitabine sensitivity was investigated in a panel of cell lines, characterized by differences in levels of activated PKB. Suppression of PKB activity was achieved with an inhibitor of phosphatidylinositol 3-kinase (wortmannin) and silencing RNA. RESULTS: Enhanced gemcitabine (2',2'-difluoro-2'-deoxycytidine)-induced cytotoxicity in vitro was achieved with suppression of high PKB activity with wortmannin in BxPC-3, PK-1, and PK-8 cells and silencing RNA targeted to total PKB, rather than PKBbeta, in PANC-1 cells. Opposite to gemcitabine sensitivity levels in vitro, the growth of PANC-1 xenografts was inhibited with gemcitabine treatment, whereas BxPC-3 became drug resistant. Monolayer cell cultures reestablished from solid tumors behaved similarly to original cultures, suggesting that the tumor microenvironment has a critical role in determining drug sensitivity. A comparison of transcript profiles of the models indicated that PKB signaling might be modulated by a number of pathways responsive to the tumor hypoxia microenvironment. CONCLUSIONS: These results suggested that gemcitabine efficacy involving the PKB pathway depends on PKB activity, its mechanisms of enhanced activity, as well as its function in a signaling network.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Carcinoma, Pancreatic Ductal/enzymology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/physiology , Neoplasm Proteins/physiology , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Androstadienes/pharmacology , Animals , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Hypoxia , Cell Line, Tumor/enzymology , Cell Line, Tumor/transplantation , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Gene Expression Profiling , Humans , Male , Mice , Mice, SCID , Neoplasm Proteins/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/pharmacology , Wortmannin , Xenograft Model Antitumor Assays , Gemcitabine
12.
Biochem J ; 372(Pt 1): 53-64, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12603202

ABSTRACT

We have previously reported three Caenorhabditis elegans genes ( gly-12, gly-13 and gly-14 ) encoding UDP- N -acetyl-D-glucosamine:alpha-3-D-mannoside beta1,2- N -acetylglucosaminyltransferase I (GnT I), an enzyme essential for hybrid and complex N-glycan synthesis. GLY-13 was shown to be the major GnT I in worms and to be the only GnT I cloned to date which can act on [Manalpha1,6(Manalpha1,3)Manalpha1,6](Manalpha1,3)Manbeta1, 4GlcNAcbeta1,4GlcNAc-R, but not on Manalpha1,6(Manalpha1,3)Manbeta1- O -R substrates. We now report the kinetic constants, bivalent-metal-ion requirements, and optimal pH, temperature and Mn(2+) concentration for this unusual enzyme. C. elegans glycoproteins are rich in oligomannose (Man(6-9)GlcNAc(2)) and 'paucimannose' Man(3-5)GlcNAc(2)(+/-Fuc) N-glycans, but contain only small amounts of complex and hybrid N-glycans. We show that the synthesis of paucimannose Man(3)GlcNAc(2) requires the prior actions of GnT I, alpha3,6-mannosidase II and a membrane-bound beta- N -acetylglucosaminidase similar to an enzyme previously reported in insects. The beta- N -acetylglucosaminidase removes terminal N -acetyl-D-glucosamine from the GlcNAcbeta1, 2Manalpha1,3Manbeta- arm of Manalpha1,6(GlcNAcbeta1,2Manalpha1,3) Manbeta1,4GlcNAcbeta1,4GlcNAc-R to produce paucimannose Man(3)GlcNAc(2) N-glycan. N -acetyl-D-glucosamine removal was inhibited by two N -acetylglucosaminidase inhibitors. Terminal GlcNAc was not released from [Manalpha1,6(Manalpha1,3)Manalpha 1,6] (GlcNAcbeta1,2Manalpha1,3)Manbeta1,4GlcNAcbeta1,4GlcNAc-R nor from the GlcNAcbeta1,2Manalpha1,6Manbeta- arm. These findings indicate that GLY-13 plays an important role in the synthesis of N-glycans by C. elegans and that therefore the worm should prove to be a suitable model for the study of the role of GnT I in nematode development.


Subject(s)
Acetylglucosaminidase/metabolism , Caenorhabditis elegans/metabolism , Mannosidases/metabolism , N-Acetylglucosaminyltransferases/metabolism , Polysaccharides/biosynthesis , Animals , Caenorhabditis elegans/enzymology , Kinetics , Microsomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL