Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Orphanet J Rare Dis ; 19(1): 9, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38183136

ABSTRACT

BACKGROUND: Ehlers-Danlos syndrome Type IV (aka Vascular Ehlers Danlos, or vEDS) is a dominantly inherited mutation in the Collagen 3A1 gene (COL3A1). The disease is characterized by tissue friability and age-related susceptibility to arterial aneurysm, dissection and rupture as well as uterine and bowl tears. These clinical manifestations result in major surgical intervention and decreased life expectancy. Understanding how mutations in COL3A1 impact the structure and function of the extracellular matrix (ECM) is important to managing the disease and finding treatments. RESULTS: Skin fibroblasts from vEDS subjects heterozygous for the p.G588S pathogenic variant in the COL3A1 gene and a normal individual were cultured and studied. Proteomics analysis identified dozens of upregulated proteins related to extracellular matrix dysregulation that is characteristic of fibrosis. Gene expression libraries from cultured primary fibroblasts were screened for messenger RNA (mRNA) markers of ECM degradation. The proteomics and targeted gene expression array results were largely consistent with dysregulation of the extracellular matrix in vEDS. The data show upregulation of multiple Collagen proteins and genes, other ECM components, and enzymes related to ECM processing and turn-over. vEDS fibroblasts expressed significantly more cross linked C-Telopeptide of Collagen III (CTXIII) than normal fibroblasts, indicative of Collagen III degradation and turn-over. Further, the expression and activity of Lysyl Oxidase (LOX), an enzyme that initiates covalent cross-linking of soluble collagen and elastin into protease resistant fibers, is elevated in vEDS fibroblasts compared to normal fibroblasts. CONCLUSION: Together, these findings suggest dysregulated ECM deposition and processing, reminiscent of a state of fibrosis. Therapeutics that target the dysregulated ECM proteins or help replace damaged tissue may improve clinical outcomes.


Subject(s)
Ehlers-Danlos Syndrome , Protein-Lysine 6-Oxidase , Humans , Protein-Lysine 6-Oxidase/genetics , Skin , Extracellular Matrix , Fibrosis , Ehlers-Danlos Syndrome/genetics
2.
Future Sci OA ; 1(4): FSO76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-28031925

ABSTRACT

AIM: A bioanalytical method using inductively-coupled plasma-mass spectrometry to measure endogenous levels of strontium in human serum was developed and validated. RESULTS & METHODOLOGY: This article details the experimental procedures used for the method development and validation thus demonstrating the application of the inductively-coupled plasma-mass spectrometry method for quantification of strontium in human serum samples. The assay was validated for specificity, linearity, accuracy, precision, recovery and stability. Significant endogenous levels of strontium are present in human serum samples ranging from 19 to 96 ng/ml with a mean of 34.6 ± 15.2 ng/ml (SD). DISCUSSION & CONCLUSION: Calibration procedures and sample pretreatment were simplified for high throughput analysis. The validation demonstrates that the method was sensitive, selective for quantification of strontium (88Sr) and is suitable for routine clinical testing of strontium in human serum samples.

3.
Orphanet J Rare Dis ; 6: 84, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22176730

ABSTRACT

BACKGROUND: This study was conducted to identify potential biomarkers that could be used to evaluate disease progression and monitor responses to enzyme replacement therapy (ERT) in patients with mucopolysaccharidosis (MPS) IVA. METHODS: Levels of 88 candidate biomarkers were compared in plasma samples from 50 healthy controls and 78 MPSIVA patients not receiving ERT to test for significant correlations to the presence of MPSIVA. MPSIVA samples were also tested for correlations between candidate biomarkers and age, endurance, or urinary keratin sulfate (KS) levels. Then, levels of the same 88 analytes were followed over 36 weeks in 20 MPSIVA patients receiving ERT to test for significant correlations related to ERT, age, or endurance. RESULTS: Nineteen candidate biomarkers were significantly different between MPSIVA and unaffected individuals. Of these, five also changed significantly in response to ERT: alpha-1-antitrypsin, eotaxin, lipoprotein(a), matrix metalloprotein (MMP)-2, and serum amyloid P. Three of these were significantly lower in MPSIVA individuals versus unaffected controls and were increased during ERT: alpha-1-antitrypsin, lipoprotein(a), and serum amyloid P. CONCLUSIONS: Candidate biomarkers alpha-1-antitrypsin, lipoprotein(a), and serum amyloid P may be suitable markers, in addition to urinary KS, to follow the response to ERT in MPSIVA patients.


Subject(s)
Biomarkers/blood , Biomarkers/urine , Chondroitinsulfatases/administration & dosage , Enzyme Replacement Therapy/methods , Mucopolysaccharidoses/therapy , Mucopolysaccharidosis IV/physiopathology , Mucopolysaccharidosis IV/therapy , Adult , Aged , Chondroitinsulfatases/therapeutic use , Disease Progression , Dose-Response Relationship, Drug , Enzyme Replacement Therapy/adverse effects , Female , Humans , Keratins/urine , Lipoprotein(a)/blood , Male , Middle Aged , Mucopolysaccharidoses/physiopathology , Serum Amyloid P-Component/metabolism , Treatment Outcome , Young Adult , alpha 1-Antitrypsin/blood
4.
Bioanalysis ; 3(16): 1855-66, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21877895

ABSTRACT

BACKGROUND: Mucopolysaccharidosis IVA (MPS IVA, Morquio A syndrome) is an inherited lysosomal storage disease caused by deficiency of N-acetylgalactosamine-6-sulfatase (GALNS), an enzyme required for stepwise degradation of keratan sulfate (KS). We have developed a selective, sensitive, accurate and precise LC-MS/MS assay for the KS-derived disaccharides Galß1-4GlcNAc(6S) and Gal(6S)ß1-4GlcNAc(6S) in human urine and plasma using keratanase II digestion. RESULTS: Mean accuracy was 96-106% in urine and 97-108% in plasma. Precision was high, with relative standard deviations of 1-2% (intra-day) and 2-5% (inter-day) in urine and 1-2% (intra-day) and 4-7% (inter-day) in plasma. The lower limit of quantitation was 0.026 µg/ml (plasma) and 0.104 µg/ml (urine), with a quantitation range of 0.026-5 µg/ml (plasma) and 0.104-20 µg/ml (urine). CONCLUSION: Clinical sample analysis in 168 MPS IVA patients and 225 healthy controls demonstrates the clinical utility of this method.


Subject(s)
Chromatography, Liquid/methods , Disaccharides/blood , Disaccharides/urine , Keratan Sulfate/blood , Keratan Sulfate/urine , Mucopolysaccharidosis IV/diagnosis , Adolescent , Adult , Biomarkers/blood , Biomarkers/urine , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Mass Spectrometry/methods , Middle Aged , Mucopolysaccharidosis IV/blood , Mucopolysaccharidosis IV/urine
5.
Clin Ther ; 32(2): 338-46, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20206791

ABSTRACT

BACKGROUND: Phenylketonuria (PKU) is an autosomal recessive metabolic disorder characterized by hyperphenylalaninemia in association with neurocognitive and neuromotor impairment. Sapropterin dihydrochloride (hereafter referred to as sapropterin) administered orally as dissolved tablets is approved by the US Food and Drug Administration for hyperphenylalaninemia in patients with tetrahydrobiopterin responsive PKU. OBJECTIVES: This study compared the relative oral bioavailability of sapropterin when administered as intact and dissolved tablets. It also assessed the effect of food on the oral bioavailability of sapropterin administered as intact tablets. METHODS: This was a randomized, open-label, 3-treatment, 6-sequence, 3-period crossover study in healthy male and female subjects. Subjects were randomized to receive single oral 10-mg/kg doses of sapropterin administered as dissolved tablets after a fast; as intact tablets after a fast; and as intact tablets with a high-calorie, high-fat meal. The 3 dosing periods were separated by a washout period of at least 7 days. In each dosing period, blood samples were obtained within 40 minutes before and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 18, and 24 hours after dosing. A follow-up assessment was performed 5 to 7 days after the last dosing period. The relative bioavailability of sapropterin from the 3 dosing regimens was assessed based on C(max), AUC(0-t), and AUC(0-infinity), estimated from calculated plasma tetrahydrobiopterin concentrations using a noncompartmental model. Safety assessments included physical examinations, clinical laboratory tests, and ECGs at the beginning and end of the study. Vital signs were monitored periodically during each treatment period. RESULTS: The study enrolled 32 healthy subjects (16 men, 16 women) with a mean (SD) age of 29.2 (9.0) years, height of 172.7 (10.0) cm, weight of 73.0 (13.9) kg, and body mass index ranging from 18 to 30 kg/m(2). Twenty-three were white, 5 African American, 2 Asian/Pacific Islander, 1 Hispanic, and 1 Native American. The estimated geometric mean ratio of AUC(0-t) for intact compared with dissolved tablets under fasting conditions was 141.24% (90% CI, 122.05-163.43), and the geometric mean ratio of AUC(0-t) for intact tablets under fed compared with fasting conditions was 143.46% (90% CI, 124.22-165.69). Nine subjects (28.1%) reported a total of 20 treatment-emergent adverse events (AEs). The most frequently reported AEs were gastrointestinal disorders (6 subjects [18.8%]) and central nervous system disorders (4 [12.5%]). Eight AEs considered possibly or probably related to sapropterin were reported by 4 subjects (12.5%); these were of mild severity and gastrointestinal in nature. No severe or serious AEs or discontinuations due to AEs occurred during the study. CONCLUSIONS: Administration of sapropterin as intact tablets and with a high-calorie, high-fat meal was associated with increased drug exposure. Oral administration of sapropterin 10 mg/kg as intact tablets with or without food was generally well tolerated.


Subject(s)
Biopterins/analogs & derivatives , Dietary Fats/administration & dosage , Food-Drug Interactions , Administration, Oral , Adolescent , Adult , Area Under Curve , Biological Availability , Biopterins/administration & dosage , Biopterins/blood , Biopterins/pharmacokinetics , Cross-Over Studies , Energy Intake , Fasting , Female , Humans , Male , Middle Aged , Models, Biological , Postprandial Period , Solubility , Tablets , Young Adult
6.
Bioanalysis ; 1(5): 895-903, 2009 Aug.
Article in English | MEDLINE | ID: mdl-21083061

ABSTRACT

BACKGROUND: Tetrahydrobiopterin (BH4) is a naturally occurring pteridine and cofactor for a variety of enzymes, including phenylalanine-4-hydroxylase, nitric oxide synthetase and glyceryl ether monooxygenase. BH4 is readily oxidized to dihydrobiopterin and biopterin (B), however only BH4 can provide proper cofactor functions. BH4 is the active ingredient in Kuvan™ for the treatment of phenylketonuria. In order to measure BH4 in plasma from nonclinical and clinical samples with good accuracy, precision, sensitivity and robustness, an LC-MS/MS method was validated. To overcome the oxidation of BH4 in postcollection plasma, the approach was to measure the concentration of BH4 indirectly by measuring B concentration and applying an oxidation conversion ratio. Different endogenous levels of BH4 are determined in human, monkey, dog, rabbit, rat and mouse plasma. Furthermore, the conversion ratio of BH4 to B for each species is different and determined empirically. Plasma is transferred into cryogenic vials containing 0.1% dithioerythritol to prevent oxidation of BH4. The samples are then extracted and oxidized under basic conditions. B is measured with LC-MS/MS using negative ion mode. RESULTS: The method is accurate, and precise to within 15%. The lower limit of quantitation in matrix is 5, 50 or 100 ng/ml, depending on the species endogenous levels of BH4. The pharmacokinetics of a single oral dose at three concentrations of BH4 administered to C57BL/6 mice is presented. In this mouse study, the T(1/2) of BH4 in plasma was approximately 1.2 h. CONCLUSION: The validated LC-MS/MS method to determine plasma BH4 concentration described herein has been used to support many nonclinical and clinical toxicokinetic and pharmacokinetic studies. BH4 is sensitive to oxidation and has a complicated biology. The method successfully supported the approval of Kuvan for the treatment of phenylketonuria.


Subject(s)
Biopterins/analogs & derivatives , Biopterins/blood , Chromatography, High Pressure Liquid/methods , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Animals , Biopterins/pharmacokinetics , Humans , Least-Squares Analysis , Mice , Mice, Inbred C57BL , Phenylketonurias/drug therapy , Reproducibility of Results
7.
Clin Pharmacokinet ; 47(12): 817-25, 2008.
Article in English | MEDLINE | ID: mdl-19026037

ABSTRACT

BACKGROUND AND OBJECTIVE: Untreated phenylketonuria is characterized by neurocognitive and neuromotor impairment, which result from elevated blood phenylalanine concentrations. To date, the recommended management of phenylketonuria has been the use of a protein-restricted diet and the inclusion of phenylalanine-free protein supplements; however, this approach is often associated with poor compliance and a suboptimal clinical outcome. Sapropterin dihydrochloride, herein referred to as sapropterin, a synthetic formulation of 6R-tetrahydrobiopterin (6R-BH4), has been shown to be effective in reducing blood phenylalanine concentrations in patients with phenylketonuria. The objective of the current study was to characterize the pharmacokinetics and pharmacokinetic variability of sapropterin and to identify the characteristics that influence this variability. PATIENTS AND METHODS: This was a 12-week, fixed-dose phase of an open-label extension study. The study was conducted at 26 centres in North America and Europe.Patients with phenylketonuria were eligible to participate if they were > or =8 years of age and had received > or =80% of the scheduled doses in a previous 6-week, randomized, placebo-controlled study or had been withdrawn from that study after exceeding a plasma phenylalanine concentration of > or =1500 micromol/L to > or =1800 micromol/L, depending on the subject's age and baseline plasma phenylalanine concentration. A total of 78 patients participated. Patients received oral once-daily doses of sapropterin (Kuvan) 5, 10 or 20 mg/kg/day. Blood samples for the pharmacokinetic analysis were obtained during weeks 6, 10 and 12. A D-optimal sparse sampling strategy was used, and data were analysed by population-based, nonlinear, mixed-effects modelling methods. MAIN OUTCOME MEASURE: In a prospectively planned analysis, the apparent clearance, apparent volume of distribution, absorption rate constant and associated interindividual variabilities of each parameter were estimated by modelling observed BH4 plasma concentration-time data. RESULTS: The best structural model to describe the pharmacokinetics of sapropterin was a two-compartment model with first-order input, first-order elimination and a baseline endogenous BH4 concentration term. Total bodyweight was the only significant covariate identified, the inclusion of which on both the apparent clearance (mean = 2100 L/h/70 kg) and central volume of distribution (mean = 8350 L/70 kg) substantially improved the model's ability to describe the data. The mean (SD) terminal half-life of sapropterin was 6.69 (2.29) hours and there was little evidence of accumulation, even at the highest dose. CONCLUSION: These findings, taken together with the observed therapeutic effect, support bodyweight-based, once-daily dosing of sapropterin 5-20 mg/kg/day.


Subject(s)
Biopterins/analogs & derivatives , Phenylketonurias/drug therapy , Administration, Oral , Adolescent , Adult , Biological Availability , Biopterins/administration & dosage , Biopterins/pharmacokinetics , Body Weight/drug effects , Child , Creatinine/blood , Dose-Response Relationship, Drug , Europe , Female , Follow-Up Studies , Half-Life , Humans , Male , Metabolic Clearance Rate , Models, Biological , Phenylalanine/blood , Phenylketonurias/blood , United States , Young Adult
8.
AAPS J ; 10(3): 439-49, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18709516

ABSTRACT

Most patients receiving Naglazyme (galsulfase, rhASB) enzyme replacement therapy for mucopolysaccharidosis type VI develop an antibody response. To evaluate the impact of this response, two in vitro neutralizing antibody (NAb) assays were developed based on the two steps of the mechanism of action. Neutralization of enzyme activity was detected by inhibition of rhASB cleavage of a fluorogenic substrate. Neutralization of receptor binding was detected by decreased binding of labeled rhASB to immobilized soluble receptor. For the enzyme activity NAb assay, serum pretreatment was required to isolate antibodies from interfering phosphate ions, with sensitivity of < or =5 microg/mL. The receptor binding NAb assay used a five-fold dilution, with sensitivity of < or =40 microg/mL. Cutpoints for percent inhibition were based on 95% confidence intervals from naïve sera. Clinical samples were similarly likely to be positive in both assays than positive for neutralization of only one step in the mechanism of action. The two NAb assays yielded complementary information about potential neutralization of rhASB. Relative estimated sensitivity between neutralization assays did not correlate with the number of positive clinical samples or patients. In vitro NAb assays based on a well-understood mechanism of action provide specific information about the NAb mechanism.


Subject(s)
Antibody Formation/drug effects , Enzyme-Linked Immunosorbent Assay/methods , N-Acetylgalactosamine-4-Sulfatase/adverse effects , Receptors, Cell Surface/metabolism , Antibodies/blood , Antibody Formation/immunology , Biotin/immunology , Humans , In Vitro Techniques , N-Acetylgalactosamine-4-Sulfatase/metabolism , Protein Binding , Receptor, IGF Type 2/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/metabolism , Sensitivity and Specificity
9.
AAPS J ; 10(2): 363-72, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18607760

ABSTRACT

Naglazyme (galsulfase, rhASB) was developed as enzyme replacement therapy for mucopolysaccharidosis type VI. Naglazyme generated an IgG antibody response in most patients. To better characterize Naglazyme immunogenicity, a solution phase bridged immunoassay was developed to measure total antibody response regardless of isotype. Overnight incubation of serum dilutions with rhASB labeled with biotin and ruthenium-based tags allowed antibody-antigen complexes to form prior to capture on a streptavidin plate. Neat serum was tolerated in the assay, with a 1:10 screening dilution implemented for testing. At this dilution, the assay was sensitive to 75 ng/ml anti-rhASB. Titers were reported as the highest dilution factor with signal above a 95% confidence interval from naïve individual sera. Precise measurement of titers, within two consecutive dilution factors, was observed across analysts and days. Clinical samples showed similar positive/negative results between the IgG ELISA and the total antibody ECLA, although with an imperfect correlation. Improvements in assay performance and implementation strategy altered some positive clinical samples to negative and vice versa. Comparison of the titer readout for clinical samples with the screening signal illustrates a range of relationships for signal versus sample dilution factor, confirming that signal from a screening dilution cannot directly predict the reported titer.


Subject(s)
Antibody Formation/drug effects , Luminescent Measurements/methods , N-Acetylgalactosamine-4-Sulfatase/adverse effects , Adult , Child , Enzyme-Linked Immunosorbent Assay/methods , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mucopolysaccharidosis VI/blood , Mucopolysaccharidosis VI/drug therapy , Mucopolysaccharidosis VI/immunology , Recombinant Proteins/adverse effects , Reproducibility of Results , Sensitivity and Specificity , Specimen Handling
10.
Mol Cell Biol ; 26(19): 7283-98, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16980629

ABSTRACT

Changes in synaptic plasticity required for memory formation are dynamically regulated through opposing excitatory and inhibitory neurotransmissions. To explore the potential contribution of NF-kappaB/Rel to these processes, we generated transgenic mice conditionally expressing a potent NF-kappaB/Rel inhibitor termed IkappaBalpha superrepressor (IkappaBalpha-SR). Using the prion promoter-enhancer, IkappaBalpha-SR is robustly expressed in inhibitory GABAergic interneurons and, at lower levels, in excitatory neurons but not in glia. This neuronal pattern of IkappaBalpha-SR expression leads to decreased expression of glutamate decarboxylase 65 (GAD65), the enzyme required for synthesis of the major inhibitory neurotransmitter, gamma-aminobutyric acid (GABA) in GABAergic interneurons. IkappaBalpha-SR expression also results in diminished basal GluR1 levels and impaired synaptic strength (input/output function), both of which are fully restored following activity-based task learning. Consistent with diminished GAD65-derived inhibitory tone and enhanced excitatory firing, IkappaBalpha-SR+ mice exhibit increased late-phase long-term potentiation, hyperactivity, seizures, increased exploratory activity, and enhanced spatial learning and memory. IkappaBalpha-SR+ neurons also express higher levels of the activity-regulated, cytoskeleton-associated (Arc) protein, consistent with neuronal hyperexcitability. These findings suggest that NF-kappaB/Rel transcription factors act as pivotal regulators of activity-dependent inhibitory and excitatory neuronal function regulating synaptic plasticity and memory.


Subject(s)
Long-Term Potentiation/physiology , Neuronal Plasticity , Neurons/metabolism , Synapses/metabolism , Transcription Factor RelA/metabolism , Animals , Behavior, Animal , Cognition/physiology , Gene Expression Regulation, Enzymologic , Glutamate Decarboxylase/genetics , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/pathology , I-kappa B Proteins/metabolism , Isoenzymes/genetics , Memory/physiology , Mice , Mice, Transgenic , NF-KappaB Inhibitor alpha , Neurons/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, AMPA/metabolism , Synaptic Transmission/physiology , Transcription Factor RelA/antagonists & inhibitors , gamma-Aminobutyric Acid/metabolism
11.
Cancer Res ; 66(4): 2271-8, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16489031

ABSTRACT

The receptor protein tyrosine phosphatase beta (RPTPbeta) is a functional biomarker for several solid tumor types. RPTPbeta expression is largely restricted to the central nervous system and overexpressed primarily in astrocytic tumors. RPTPbeta is known to facilitate tumor cell adhesion and migration through interactions with extracellular matrix components and the growth factor pleiotrophin. Here, we show that RPTPbeta is expressed in a variety of solid tumor types with low expression in normal tissue. To assess RPTPbeta as a potential target for treatment of glioblastoma and other cancers, antibodies directed to RPTPbeta have been developed and profiled in vitro and in vivo. The recombinant extracellular domain of human short RPTPbeta was used to immunize mice and generate monoclonal antibodies that selectively recognize RPTPbeta and bind to the antigen with low nanomolar affinities. Moreover, these antibodies recognized the target on living tumor cells as measured by flow cytometry. These antibodies killed glioma cells in vitro when coupled to the cytotoxin saporin either directly or via a secondary antibody. Finally, in vivo studies showed that an anti-RPTPbeta immunotoxin (7E4B11-SAP) could significantly delay human U87 glioma tumors in a mouse xenograft model. Unconjugated 7E4B11 provides a modest but statistically significant tumor growth delay when delivered systemically in mice bearing U87 glioma tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Glioblastoma/enzymology , Glioblastoma/therapy , Nerve Tissue Proteins/immunology , Protein Tyrosine Phosphatases/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunization , Immunotoxins/immunology , Immunotoxins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Nerve Tissue Proteins/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Xenograft Model Antitumor Assays
12.
J Neurooncol ; 76(3): 239-48, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16234985

ABSTRACT

Invasion of glioma cells involves the attachment of invading tumor cells to extracellular matrix (ECM), disruption of ECM components, and subsequent cell penetration into adjacent brain structures. Discoidin domain receptor 1 (DDR1) tyrosine kinases constitute a novel family of receptors characterized by a unique structure in the ectodomain (discoidin-I domain). These cell surface receptors bind to several collagens and facilitate cell adhesion. Little is known about DDR1 expression and function in glioblastoma multiforme. In this study we demonstrate that DDR1 is overexpressed in glioma tissues using cDNA arrays, immunohistochemistry and Western blot analysis. Functional comparison of two splice variants of DDR1 (DDR1a and DDR1b) reveal novel differences in cell based glioma models. Overexpression of either DDR1a or DDR1b caused increased cell attachment. However, glioma cells overexpressing DDR1a display enhanced invasion and migration. We also detect increased levels of matrix metalloproteinase-2 in DDR1a overexpressing cells as measured by zymography. Inhibition of MMP activity using MMP inhibitor suppressed DDR1a stimulated cell-invasion. Similarly, an antibody against DDR1 reduced DDR1a mediated invasion as well as the enhanced adhesion of DDR1a and DDR1b overexpressing cells. These results suggest that DDR1a plays a critical role in inducing tumor cell adhesion and invasion, and this invasive phenotype is caused by activation of matrix metalloproteinase-2.


Subject(s)
Brain Neoplasms/pathology , Cell Adhesion/physiology , Glioma/pathology , Matrix Metalloproteinase 2/metabolism , Neoplasm Invasiveness , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Blotting, Western , Brain Neoplasms/metabolism , Cell Line, Tumor , Discoidin Domain Receptors , Enzyme Activation/physiology , Glioma/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Oligonucleotide Array Sequence Analysis , Protein Isoforms/metabolism , Transfection
13.
J Neurooncol ; 74(3): 241-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16187021

ABSTRACT

Astrocytomas are the most common primary tumor of the adult human central nervous system. Despite efforts to develop more effective clinical treatment strategies, median survival time for patients with the most severe form of astrocytoma, glioblastoma multiforme (GBM), remains about one year. Astrocytomas are resistant to cytotoxic therapy in general and radiation therapy in particular, greatly limiting treatment options. One reason for this seems to be defects in the pathways controlling apoptosis. We have characterized the role of the tyrosine phosphatase FAP-1 (FAS-associated phosphatase 1) in astrocytomas. Our studies demonstrate that FAP-1 is overexpressed in astrocytomas and this contributes to the resistance of the tumor cells to FAS-mediated apoptosis. We demonstrate that knockdown of FAP-1 by RNA interference leads to increased apoptosis and increased sensitivity of astrocytoma cells to FAS-induced cell death. FAP-1 binds to FAS in a ligand-dependent manner and forms a signaling complex that modulates the ability of astrocytoma cells to undergo FAS ligand (FASL)-mediated cell death. In astrocytoma cells, FASL treatment induces tyrosine phosphorylation of FAS. FAP-1 dephosphorylates phospho-tyrosine 275 in the carboxyl terminus of FAS. This is the first direct evidence that FAS activity can be regulated by reversible phosphorylation and suggests a mechanism for astrocytoma resistance to apoptosis.


Subject(s)
Apoptosis/physiology , Astrocytoma/pathology , Brain Neoplasms/pathology , Protein Tyrosine Phosphatases/metabolism , fas Receptor/metabolism , Cell Line, Tumor , Gene Expression , Gene Expression Profiling , Humans , Immunoblotting , Immunohistochemistry , Phosphorylation , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 13 , Protein Tyrosine Phosphatases/analysis , Protein Tyrosine Phosphatases/genetics , RNA Interference , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
14.
Neuro Oncol ; 7(2): 154-63, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15831233

ABSTRACT

The receptor protein tyrosine phosphatase beta (RPTPbeta/PTPzeta) is overexpressed in glioblastoma tumors and plays a functional role in tumor cell migration and adhesion. Glioblastomas express at least three splice variants of RPTPbeta, including long and short receptor forms and a secreted chondroitin sulfate proteoglycan called phosphacan. Here we explore the differences in the expression pattern and function of long RPTPbeta and short RPTPbeta. The short form of RPTPbeta lacks exon 12, which encodes 860 amino acids located in the extracellular domain. Until now, functional differences between long and short RPTPbeta have been difficult to elucidate. In this study, antibodies specific to the splice junction, unique to short RPTPbeta, allowed for the discrimination of the two receptors. A study of normal brain tissue and graded astrocytomas indicates that long and short RPTPbeta forms have an overlapping expression pattern. In order to study functional differences between long and short RPTPbeta, we created stable U87 glioblastoma cells that expressed these receptors. U87 stable cell lines overexpressing long or short RPTPbeta migrate faster and adhere more robustly than parental U87 cells. The two forms differ in that long-RPTPbeta-overexpressing cells migrate and adhere better than short-RPTPbeta-overexpressing cells. A study of the extracellular domain of short RPTPbeta indicates that it retains much of the functional capacity of phosphacan. Indeed, the action of recombinant, short-RPTPbeta extracellular domain protein is similar to that of phosphacan as a repulsive substrate for glioblastoma cells. Comparison of the signaling capacity of long RPTPbeta to that of short RPTPbeta reveals very similar abilities to activate transcription pathways. Moreover, transient transfection with either long or short RPTPbeta activates NF-kappaB reporter gene transcription. Because of their tumor-restricted and largely overlapping expression patterns in glioblastoma, both RPTPbeta splice forms are potential therapeutic targets. The involvement of long and short RPTPbeta in glioma tumor cell biology also contributes to the value of RPTPbeta as a cancer target.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Isoforms/genetics , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Brain/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Humans , Immunoblotting , Immunohistochemistry , Nerve Tissue Proteins/chemistry , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Tyrosine Phosphatases/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 5
15.
Oncogene ; 24(10): 1673-82, 2005 Mar 03.
Article in English | MEDLINE | ID: mdl-15674329

ABSTRACT

GPR56 (also known as TM7XN1) is a newly discovered orphan G-protein-coupled receptor (GPCR) of the secretin family that has a role in the development of neural progenitor cells and has been linked to developmental malformations of the human brain. GPR56 diverges from other secretin-like family members in that it has an extremely large N-terminal extracellular region (381 amino acids) and contains a novel feature among this new subclass, consisting of four cysteine residues that define a GPCR proteolytic site (GPS motif) located just before the first transmembrane spanning domain. The rest of the amino-terminal domain contains a large number of possible N- and O-linked glycosylation sites similar to mucin-like proteins. These features suggest a role in cell-cell, or cell-matrix interactions. Here, we demonstrate upregulation of GPR56 in glioblastoma multiforme tumors using functional genomics. Immunohistochemistry studies confirmed the expression of GPR56 protein in a majority of glioblastoma/astrocytoma tumor samples with undetectable levels of expression in normal adult brain tissue. Immunofluorescence analysis of human glioma cells using anti-GPR56 antibodies demonstrate that GPR56 is expressed on the leading edge of membrane filopodia and colocalizes with alpha-actinin. Purified recombinant GPR56 extracellular domain protein inhibits glioma cell adhesion and causes abnormal cytoskeletal morphology and cell rounding. These results indicate that the extracellular domain may compete for unidentified ligand(s), and block the normal function of GPR56 in cell attachment. In reporter assays, overexpression of GPR56 activates the NF-kappaB, PAI-1 and TCF transcriptional response elements. These pathways have been implicated in cytoskeletal signaling, adhesion and tumor biology. The above results indicate that GPR56 serves as an adhesion GPCR and is involved in adhesion signaling.


Subject(s)
Cell Adhesion , Glioblastoma/pathology , Glioma/metabolism , Receptors, G-Protein-Coupled/physiology , Cell Line , Glioma/pathology , Humans , Immunohistochemistry , RNA, Messenger/analysis , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...