23 results on '"Zondag G"'
Search Results
2. Pyroptosis-inducing active caspase-1 as a genetic adjuvant in anti-cancer DNA vaccination.
- Author
-
Arakelian T, Oosterhuis K, Tondini E, Los M, Vree J, van Geldorp M, Camps M, Teunisse B, Zoutendijk I, Arens R, Zondag G, Ossendorp F, and van Bergen J
- Subjects
- Animals, Caspase 1 metabolism, Inflammation, Interleukin-1beta, Mice, Ovalbumin, Vaccination, Pyroptosis, Vaccines, DNA
- Abstract
Pyroptosis is a recently discovered form of inflammatory programmed necrosis characterized by caspase-1-mediated and gasdermin D-dependent cell death leading to the release of pro-inflammatory cytokines such as Interleukin-1 beta (IL-1β). Here, we evaluated whether pyroptosis could be exploited in DNA vaccination by incorporating a constitutively active variant of caspase-1 to the antigen-expressing DNA. In vitro, transfection with constitutively active caspase-1 DNA induced pro-IL-1β maturation and IL-1β release as well as gasdermin D-dependent cell death. To test active caspase-1 as a genetic adjuvant for the induction of antigen-specific T cell responses, mice were vaccinated intradermally with a DNA vaccine consisting of the active caspase-1 plasmid together with a plasmid encoding an ovalbumin-derived CD8 T cell epitope. Active caspase-1 accelerated and amplified antigen-specific CD8 T cell responses when administered simultaneously with the DNA vaccine at an equimolar dose. Moreover, upon challenge with melanoma cells expressing ovalbumin, mice vaccinated with the antigen vaccine adjuvanted with active caspase-1 showed significantly better survival compared to the non-adjuvanted group. In conclusion, we have developed a novel genetic adjuvant that for the first time employs the pyroptosis pathway to improve DNA vaccination against cancer., Competing Interests: Declaration of Competing Interest BT, GZ and JvB are employees of Immunetune. IZ and GZ are employees of Synvolux. JvB and GZ are inventors on patent application WO2020204714A1: ‘Immune-stimulatory compositions and use thereof’., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
3. Dominant Antiviral CD8 + T Cell Responses Empower Prophylactic Antibody-Eliciting Vaccines Against Cytomegalovirus.
- Author
-
Pardieck IN, van Duikeren S, Veerkamp DMB, Brasem DJ, Redeker A, van Bergen J, Han W, Ossendorp F, Zondag G, and Arens R
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Cytomegalovirus Infections immunology, Epitopes immunology, Immunity, Cellular, Immunity, Humoral, Immunization, Secondary methods, Immunoglobulin G blood, Immunoglobulin G immunology, Mice, Mice, Inbred C57BL, Specific Pathogen-Free Organisms, Vaccination, Vaccines, DNA administration & dosage, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Antibodies, Viral blood, CD8-Positive T-Lymphocytes immunology, Cytomegalovirus immunology, Cytomegalovirus Infections prevention & control, Cytomegalovirus Vaccines immunology, Vaccines, DNA immunology
- Abstract
Human cytomegalovirus (HCMV) is an ubiquitous herpesvirus that can cause serious morbidity and mortality in immunocompromised or immune-immature individuals. A vaccine that induces immunity to CMV in these target populations is therefore highly needed. Previous attempts to generate efficacious CMV vaccines primarily focused on the induction of humoral immunity by eliciting neutralizing antibodies. Current insights encourage that a protective immune response to HCMV might benefit from the induction of virus-specific T cells. Whether addition of antiviral T cell responses enhances the protection by antibody-eliciting vaccines is however unclear. Here, we assessed this query in mouse CMV (MCMV) infection models by developing synthetic vaccines with humoral immunity potential, and deliberately adding antiviral CD8
+ T cells. To induce antibodies against MCMV, we developed a DNA vaccine encoding either full-length, membrane bound glycoprotein B (gB) or a secreted variant lacking the transmembrane and intracellular domain (secreted (s)gB). Intradermal immunization with an increasing dose schedule of sgB and booster immunization provided robust viral-specific IgG responses and viral control. Combined vaccination of the sgB DNA vaccine with synthetic long peptides (SLP)-vaccines encoding MHC class I-restricted CMV epitopes, which elicit exclusively CD8+ T cell responses, significantly enhanced antiviral immunity. Thus, the combination of antibody and CD8+ T cell-eliciting vaccines provides a collaborative improvement of humoral and cellular immunity enabling enhanced protection against CMV., Competing Interests: WH, JvB, and GZ are employees of Immunetune BV. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Pardieck, van Duikeren, Veerkamp, Brasem, Redeker, van Bergen, Han, Ossendorp, Zondag and Arens.)- Published
- 2022
- Full Text
- View/download PDF
4. A poly-neoantigen DNA vaccine synergizes with PD-1 blockade to induce T cell-mediated tumor control.
- Author
-
Tondini E, Arakelian T, Oosterhuis K, Camps M, van Duikeren S, Han W, Arens R, Zondag G, van Bergen J, and Ossendorp F
- Abstract
The combination of immune-stimulating strategies has the potency to improve immunotherapy of cancer. Vaccination against neoepitopes derived from patient tumor material can generate tumor-specific T cell immunity, which could reinforce the efficacy of checkpoint inhibitor therapies such as anti-PD-1 treatment. DNA vaccination is a versatile platform that allows the inclusion of multiple neoantigen-coding sequences in a single formulation and therefore represents an ideal platform for neoantigen vaccination. We developed an anti-tumor vaccine based on a synthetic DNA vector designed to contain multiple cancer-specific epitopes in tandem. The DNA vector encoded a fusion gene consisting of three neoepitopes derived from the mouse colorectal tumor MC38 and their natural flanking sequences as 40 amino acid stretches. In addition, we incorporated as reporter epitopes the helper and CTL epitope sequences of ovalbumin. The poly-neoantigen DNA vaccine elicited T cell responses to all three neoantigens and induced functional CD8 and CD4 T cell responses to the reporter antigen ovalbumin after intradermal injection in mice. The DNA vaccine was effective in preventing outgrowth of B16 melanoma expressing ovalbumin in a prophylactic setting. Moreover, the combination of therapeutic DNA vaccination and anti-PD-1 treatment was synergistic in controlling MC38 tumor growth whereas individual treatments did not succeed. These data demonstrate the potential of DNA vaccination to target multiple neoepitopes in a single formulation and highlight the cooperation between vaccine-based and checkpoint blockade immunotherapies for the successful eradication of established tumors., (© 2019 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2019
- Full Text
- View/download PDF
5. MARK-AGE population: From the human model to new insights.
- Author
-
Capri M, Moreno-Villanueva M, Cevenini E, Pini E, Scurti M, Borelli V, Palmas MG, Zoli M, Schön C, Siepelmeyer A, Bernhardt J, Fiegl S, Zondag G, de Craen AJ, Hervonen A, Hurme M, Sikora E, Gonos ES, Voutetakis K, Toussaint O, Debacq-Chainiaux F, Grubeck-Loebenstein B, Bürkle A, and Franceschi C
- Subjects
- Animals, Humans, Aging physiology, Models, Biological
- Published
- 2015
- Full Text
- View/download PDF
6. MARK-AGE biomarkers of ageing.
- Author
-
Bürkle A, Moreno-Villanueva M, Bernhard J, Blasco M, Zondag G, Hoeijmakers JH, Toussaint O, Grubeck-Loebenstein B, Mocchegiani E, Collino S, Gonos ES, Sikora E, Gradinaru D, Dollé M, Salmon M, Kristensen P, Griffiths HR, Libert C, Grune T, Breusing N, Simm A, Franceschi C, Capri M, Talbot D, Caiafa P, Friguet B, Slagboom PE, Hervonen A, Hurme M, and Aspinall R
- Subjects
- European Union, Female, Humans, Male, Aging metabolism, Biomarkers metabolism
- Abstract
Many candidate biomarkers of human ageing have been proposed in the scientific literature but in all cases their variability in cross-sectional studies is considerable, and therefore no single measurement has proven to serve a useful marker to determine, on its own, biological age. A plausible reason for this is the intrinsic multi-causal and multi-system nature of the ageing process. The recently completed MARK-AGE study was a large-scale integrated project supported by the European Commission. The major aim of this project was to conduct a population study comprising about 3200 subjects in order to identify a set of biomarkers of ageing which, as a combination of parameters with appropriate weighting, would measure biological age better than any marker in isolation., (Copyright © 2015 The Authors. Published by Elsevier Ireland Ltd.. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
7. Age-related motor neuron degeneration in DNA repair-deficient Ercc1 mice.
- Author
-
de Waard MC, van der Pluijm I, Zuiderveen Borgesius N, Comley LH, Haasdijk ED, Rijksen Y, Ridwan Y, Zondag G, Hoeijmakers JH, Elgersma Y, Gillingwater TH, and Jaarsma D
- Subjects
- Activating Transcription Factor 3, Animals, Body Weight genetics, Bungarotoxins metabolism, Galectin 3 metabolism, Gene Expression Regulation genetics, Gliosis genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Motor Neurons metabolism, Motor Neurons ultrastructure, Muscle Strength genetics, Nerve Tissue Proteins metabolism, Neurofilament Proteins metabolism, Neuromuscular Junction metabolism, Neuromuscular Junction pathology, Reaction Time genetics, Silver Staining methods, Aging pathology, DNA-Binding Proteins deficiency, Endonucleases deficiency, Motor Neurons pathology, Nerve Degeneration genetics, Nerve Degeneration physiopathology, Spinal Cord pathology
- Abstract
Degeneration of motor neurons contributes to senescence-associated loss of muscle function and underlies human neurodegenerative conditions such as amyotrophic lateral sclerosis and spinal muscular atrophy. The identification of genetic factors contributing to motor neuron vulnerability and degenerative phenotypes in vivo are therefore important for our understanding of the neuromuscular system in health and disease. Here, we analyzed neurodegenerative abnormalities in the spinal cord of progeroid Ercc1(Delta/-) mice that are impaired in several DNA repair systems, i.e. nucleotide excision repair, interstrand crosslink repair, and double strand break repair. Ercc1(Delta/-) mice develop age-dependent motor abnormalities, and have a shortened life span of 6-7 months. Pathologically, Ercc1(Delta/-) mice develop widespread astrocytosis and microgliosis, and motor neuron loss and denervation of skeletal muscle fibers. Degenerating motor neurons in many occasions expressed genotoxic-responsive transcription factors p53 or ATF3, and in addition, displayed a range of Golgi apparatus abnormalities. Furthermore, Ercc1(Delta/-) motor neurons developed perikaryal and axonal intermediate filament abnormalities reminiscent of cytoskeletal pathology observed in aging spinal cord. Our findings support the notion that accumulation of DNA damage and genotoxic stress may contribute to neuronal aging and motor neuron vulnerability in human neuromuscular disorders.
- Published
- 2010
- Full Text
- View/download PDF
8. CE-MS for metabolic profiling of volume-limited urine samples: application to accelerated aging TTD mice.
- Author
-
Nevedomskaya E, Ramautar R, Derks R, Westbroek I, Zondag G, van der Pluijm I, Deelder AM, and Mayboroda OA
- Subjects
- Aging urine, Animals, Discriminant Analysis, Disease Models, Animal, Female, Mice, Mice, Transgenic, Multivariate Analysis, Principal Component Analysis, Tandem Mass Spectrometry, Trichothiodystrophy Syndromes metabolism, Trichothiodystrophy Syndromes urine, Electrophoresis, Capillary methods, Metabolomics methods, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization methods, Urine chemistry
- Abstract
Metabolic profiling of biological samples is increasingly used to obtain more insight into the pathophysiology of diseases. For translational studies, biological samples from animal models are explored; however, the volume of these samples can be a limiting factor for metabolic profiling studies. For instance, only a few microliters of urine is often available from small animals like mice. Hence, there is a need for a tailor-made analytical method for metabolic profiling of volume-limited samples. In the present study, the feasibility of capillary electrophoresis time-of-flight mass spectrometry (CE-ToF-MS) for metabolic profiling of urine from mice is evaluated. Special attention is paid to the analytical workflow; that is, such aspects as sample preparation, analysis, and data treatment are discussed from the metabolomics viewpoint. We show that metabolites belonging to several chemical families can be analyzed in mouse urine with the CE-ToF-MS method using minimal sample pretreatment and an in-capillary preconcentration procedure. This exemplifies the advantages of CE-ToF-MS for metabolic profiling of volume-limited samples as loss of material is minimized. The feasibility of the CE-ToF-MS-based workflow for metabolic profiling is illustrated by the analysis of urine samples from wild-type as well as from TTD mutant mice, which are a model for the accelerated aging, with osteoporosis being one of the main hallmarks.
- Published
- 2010
- Full Text
- View/download PDF
9. Metabolic profiling of accelerated aging ERCC1 d/- mice.
- Author
-
Nevedomskaya E, Meissner A, Goraler S, de Waard M, Ridwan Y, Zondag G, van der Pluijm I, Deelder AM, and Mayboroda OA
- Subjects
- Animals, DNA-Binding Proteins genetics, Disease Models, Animal, Endonucleases genetics, Mice, Multivariate Analysis, Mutation, Principal Component Analysis, Proteome metabolism, Serum chemistry, Urine chemistry, Aging, Premature metabolism, DNA-Binding Proteins metabolism, Endonucleases metabolism, Metabolome, Nuclear Magnetic Resonance, Biomolecular methods
- Abstract
Aging is a fundamental biological process for which the mechanism is still largely unknown due to its complex and multifactorial nature. Animal models allow us to simplify this complexity and to study individual factors separately. As there are many causative links between DNA repair deficiency and aging, we studied the ERCC1(d/-) mouse, which has a modified ERCC1 gene, involved in the Nucleotide Excision Repair, and as a result has a premature aging phenotype. Profiling of these mice on different levels can give an insight into the mechanisms underlying the aging phenotype. In the current study, we have performed metabolic profiling of serum and urine of these mice in comparison to wild type and in relation to aging by (1)H NMR spectroscopy. Analysis of metabolic trajectories of animals from 8 to 20 weeks suggested that wild type and ERCC1(d/-) mutants have similar age-related patterns of changes; however, the difference between genotypes becomes more prominent with age. The main differences between these two genetically diverse groups of mice were found to be associated with altered lipid and energy metabolism, transition to ketosis, and attenuated functions of the liver and kidney.
- Published
- 2010
- Full Text
- View/download PDF
10. Genomic flank-sequencing of plasposon insertion sites for rapid identification of functional genes.
- Author
-
Leveau JH, Gerards S, Fritsche K, Zondag G, and van Veen JA
- Subjects
- 3' Flanking Region genetics, Base Sequence, DNA, Bacterial genetics, Gene Library, Molecular Sequence Data, Sequence Analysis, DNA, Cloning, Molecular methods, DNA Transposable Elements genetics, Gram-Negative Bacteria genetics, Mutagenesis, Insertional methods
- Abstract
Plasposons are modified mini-Tn5 transposons for random mutagenesis of Gram-negative bacteria. Their unique design allows for the rescue cloning and sequencing of DNA that flanks insertion sites in plasposon mutants. However, this process can be laborious and time-consuming, as it involves genomic DNA isolation, restriction endonuclease treatment, subsequent religation, transformation of religated DNA into an Escherichia coli host, and re-isolation as a plasmid, which is then used as a template in sequencing reactions with primers that read from the plasposon ends into the flanking DNA regions. We describe here a method that produces flanking DNA sequences directly from genomic DNA that is isolated from plasposon mutants. By eliminating the need for rescue cloning, our protocol dramatically reduces time and effort, typically by 2 to 3 working days, as well as costs associated with digestion, ligation, transformation, and plasmid isolation. Furthermore, it allows for a high-throughput automated approach to analysis of the plasposome, i.e. the collective set of plasposon insertion sites in a plasposon mutant library. We have tested the utility of genomic flank-sequencing on three plasposon mutants of the soil bacterium Collimonas fungivorans with abolished ability to degrade chitin.
- Published
- 2006
- Full Text
- View/download PDF
11. Rho family proteins in cell adhesion and cell migration.
- Author
-
Evers EE, Zondag GC, Malliri A, Price LS, ten Klooster JP, van der Kammen RA, and Collard JG
- Subjects
- Cadherins physiology, Cell Communication, Humans, Neoplasm Invasiveness physiopathology, Cell Adhesion physiology, Cell Movement physiology, Neoplasms pathology, rho GTP-Binding Proteins physiology
- Abstract
Cell migration and the regulation of cadherin-mediated homotypic cell-cell interactions are critical events during development, morphogenesis and wound healing. Aberrations in signalling pathways involved in the regulation of cell migration and cadherin-mediated cell-cell adhesion contribute to tumour invasion and metastasis. The rho family proteins, including cdc42, rac1 and rhoA, regulate signalling pathways that mediate the distinct actin cytoskeleton changes required for both cellular motility and cell-cell adhesion. Recent studies indicate that rac directly influences rho activity at the GTPase level and that the reciprocal balance between rac and rho activity can determine epithelial or mesenchymal cell morphology and migratory behaviour of epithelial (tumour) cells.
- Published
- 2000
- Full Text
- View/download PDF
12. Intramolecular interactions between the juxtamembrane domain and phosphatase domains of receptor protein-tyrosine phosphatase RPTPmu. Regulation of catalytic activity.
- Author
-
Feiken E, van Etten I, Gebbink MF, Moolenaar WH, and Zondag GC
- Subjects
- Animals, Binding Sites, COS Cells, Catalysis, Catalytic Domain, Gene Library, Models, Molecular, Peptide Fragments chemistry, Protein Conformation, Receptor-Like Protein Tyrosine Phosphatases, Class 2, Recombinant Proteins metabolism, Saccharomyces cerevisiae, Transfection, Cell Membrane enzymology, Peptide Fragments metabolism, Protein Tyrosine Phosphatases chemistry, Protein Tyrosine Phosphatases metabolism
- Abstract
RPTPmu is a receptor-like protein-tyrosine phosphatase (RPTP) whose ectodomain mediates homotypic cell-cell interactions. The intracellular part of RPTPmu contains a relatively long juxtamembrane domain (158 amino acids; aa) and two conserved phosphatase domains (C1 and C2). The membrane-proximal C1 domain is responsible for the catalytic activity of RPTPmu, whereas the membrane-distal C2 domain serves an unknown function. The regulation of RPTP activity remains poorly understood, although dimerization has been proposed as a general mechanism of inactivation. Using the yeast two-hybrid system, we find that the C1 domain binds to an N-terminal noncatalytic region in RPTPmu, termed JM (aa 803-955), consisting of a large part of the juxtamembrane domain (120 aa) and a small part of the C1 domain (33 aa). When co-expressed in COS cells, the JM polypeptide binds to both the C1 and the C2 domain. Strikingly, the isolated JM polypeptide fails to interact with either full-length RPTPmu or with truncated versions of RPTPmu that contain the JM region, consistent with the JM-C1 and JM-C2 interactions being intramolecular rather than intermolecular. Furthermore, we find that large part of the juxtamembrane domain (aa 814-922) is essential for C1 to be catalytically active. Our findings suggest a model in which RPTPmu activity is regulated by the juxtamembrane domain undergoing intramolecular interactions with both the C1 and C2 domain.
- Published
- 2000
- Full Text
- View/download PDF
13. Oncogenic Ras downregulates Rac activity, which leads to increased Rho activity and epithelial-mesenchymal transition.
- Author
-
Zondag GC, Evers EE, ten Klooster JP, Janssen L, van der Kammen RA, and Collard JG
- Subjects
- Animals, Cell Transformation, Neoplastic, Cells, Cultured, Dogs, Down-Regulation, Epithelial Cells cytology, Guanine Nucleotide Exchange Factors metabolism, Kidney cytology, Mesoderm cytology, Mitogen-Activated Protein Kinases metabolism, Models, Biological, Morphogenesis, Proteins metabolism, Proto-Oncogene Proteins c-raf metabolism, Signal Transduction, Epithelial Cells physiology, Mesoderm physiology, rac GTP-Binding Proteins metabolism, ras Proteins metabolism, rho GTP-Binding Proteins metabolism
- Abstract
Proteins of the Rho family regulate cytoskeletal rearrangements in response to receptor stimulation and are involved in the establishment and maintenance of epithelial cell morphology. We recently showed that Rac is able to downregulate Rho activity and that the reciprocal balance between Rac and Rho activity is a major determinant of cellular morphology and motility in NIH3T3 fibroblasts. Using biochemical pull-down assays, we analyzed the effect of transient and sustained oncogenic Ras signaling on the activation state of Rac and Rho in epithelial MDCK cells. In contrast to the activation of Rac by growth factor-induced Ras signaling, we found that sustained signaling by oncogenic RasV12 permanently downregulates Rac activity, which leads to upregulation of Rho activity and epithelial-mesenchymal transition. Oncogenic Ras decreases Rac activity through sustained Raf/MAP kinase signaling, which causes transcriptional downregulation of Tiam1, an activator of Rac in epithelial cells. Reconstitution of Rac activity by expression of Tiam1 or RacV12 leads to downregulation of Rho activity and restores an epithelial phenotype in mesenchymal RasV12- or RafCAAX-transformed cells. The present data reveal a novel mechanism by which oncogenic Ras is able to interfere with the balance between Rac and Rho activity to achieve morphological transformation of epithelial cells.
- Published
- 2000
- Full Text
- View/download PDF
14. Receptor protein-tyrosine phosphatase RPTPmu binds to and dephosphorylates the catenin p120(ctn).
- Author
-
Zondag GC, Reynolds AB, and Moolenaar WH
- Subjects
- Animals, Binding Sites, Catalytic Domain, Catenins, Cell Adhesion Molecules analysis, Cell Line, Cytoskeletal Proteins metabolism, Humans, Mink, Phosphoproteins analysis, Phosphorylation, Protein Tyrosine Phosphatases analysis, Tyrosine metabolism, beta Catenin, Delta Catenin, Cell Adhesion Molecules metabolism, Cell Communication, Phosphoproteins metabolism, Protein Tyrosine Phosphatases metabolism, Trans-Activators
- Abstract
RPTPmu is a prototypic receptor-like protein-tyrosine phosphatase (RPTP) that mediates homotypic cell-cell interactions. Intracellularly, RPTPmu consists of a relatively large juxtamembrane region and two phosphatase domains, but little is still known about its substrate(s). Here we show that RPTPmu associates with the catenin p120(ctn), a tyrosine kinase substrate and an interacting partner of cadherins. No interaction is detectable between RPTPmu and beta-catenin. Furthermore, we show that tyrosine-phosphorylated p120(ctn) is dephosphorylated by RPTPmu both in vitro and in intact cells. Complex formation between RPTPmu and p120(ctn) does not require tyrosine phosphorylation of p120(ctn). Mutational analysis reveals that both the juxtamembrane region and the second phosphatase domain of RPTPmu are involved in p120(ctn) binding. The RPTPmu-interacting domain of p120(ctn) maps to its unique N terminus, a region distinct from the cadherin-interacting domain. A mutant form of p120(ctn) that fails to bind cadherins can still associate with RPTPmu. Our findings indicate that RPTPmu interacts with p120(ctn) independently of cadherins, and they suggest that this interaction may serve to control the tyrosine phosphorylation state of p120(ctn) at sites of cell-cell contact.
- Published
- 2000
- Full Text
- View/download PDF
15. Acute loss of cell-cell communication caused by G protein-coupled receptors: a critical role for c-Src.
- Author
-
Postma FR, Hengeveld T, Alblas J, Giepmans BN, Zondag GC, Jalink K, and Moolenaar WH
- Subjects
- Animals, CSK Tyrosine-Protein Kinase, Cell Line, Connexin 43 metabolism, Electrodes, HeLa Cells, Humans, Mice, Patch-Clamp Techniques, Protein-Tyrosine Kinases antagonists & inhibitors, Rats, src-Family Kinases, Cell Communication physiology, GTP-Binding Proteins metabolism, Protein-Tyrosine Kinases metabolism, Receptors, Cell Surface metabolism, Signal Transduction
- Abstract
Gap junctions mediate cell-cell communication in almost all tissues, but little is known about their regulation by physiological stimuli. Using a novel single-electrode technique, together with dye coupling studies, we show that in cells expressing gap junction protein connexin43, cell-cell communication is rapidly disrupted by G protein-coupled receptor agonists, notably lysophosphatidic acid, thrombin, and neuropeptides. In the continuous presence of agonist, junctional communication fully recovers within 1-2 h of receptor stimulation. In contrast, a desensitization-defective G protein-coupled receptor mediates prolonged uncoupling, indicating that recovery of communication is controlled, at least in part, by receptor desensitization. Agonist-induced gap junction closure consistently follows inositol lipid breakdown and membrane depolarization and coincides with Rho-mediated cytoskeletal remodeling. However, we find that gap junction closure is independent of Ca2+, protein kinase C, mitogen-activated protein kinase, or membrane potential, and requires neither Rho nor Ras activation. Gap junction closure is prevented by tyrphostins, by dominant-negative c-Src, and in Src-deficient cells. Thus, G protein-coupled receptors use a Src tyrosine kinase pathway to transiently inhibit connexin43-based cell-cell communication.
- Published
- 1998
- Full Text
- View/download PDF
16. Sphingosine 1-phosphate signalling through the G-protein-coupled receptor Edg-1.
- Author
-
Zondag GC, Postma FR, Etten IV, Verlaan I, and Moolenaar WH
- Subjects
- Adenylyl Cyclase Inhibitors, Animals, COS Cells, Calcium metabolism, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cells, Cultured, Cyclic AMP metabolism, Enzyme Activation, Lysophospholipids metabolism, Nuclear Proteins physiology, Receptors, Lysophosphatidic Acid, Receptors, Lysophospholipid, Sphingosine physiology, Spodoptera, Transcription Factors physiology, Zinc Fingers, Immediate-Early Proteins physiology, Receptors, Cell Surface physiology, Receptors, G-Protein-Coupled, Signal Transduction, Sphingosine analogs & derivatives
- Abstract
Sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) are structurally related lipid mediators that act on distinct G-protein-coupled receptors to evoke similar responses, including Ca2+ mobilization, adenylate cyclase inhibition, and mitogen-activated protein (MAP) kinase activation. However, little is still known about the respective receptors. A recently cloned putative LPA receptor (Vzg-1/Edg-2) is similar to an orphan Gi-coupled receptor termed Edg-1. Here we show that expression of Edg-1 in Sf9 and COS-7 cells results in inhibition of adenylate cyclase and activation of MAP kinase (Gi-mediated), but not Ca2+ mobilization, in response to S1P. These responses are specific in that (i) S1P action is not mimicked by LPA, and (ii) Vzg-1/Edg-2 cannot substitute for Edg-1. Thus the Edg-1 receptor is capable of mediating a subset of the cellular responses to S1P.
- Published
- 1998
- Full Text
- View/download PDF
17. Receptor protein tyrosine phosphatases: involvement in cell-cell interaction and signaling.
- Author
-
Zondag GC and Moolenaar WH
- Subjects
- Animals, Cadherins physiology, Cell Adhesion physiology, Cell Communication physiology, Cell Differentiation physiology, Humans, Morphogenesis physiology, Protein Tyrosine Phosphatases physiology, Receptors, Cell Surface physiology, Signal Transduction physiology
- Abstract
Receptor protein tyrosine phosphatases (RPTPs) represent a relatively new family of cell-surface receptors consisting of a variable, putative ligand-binding ectodomain followed by a single transmembrane segment and one or two intracellular catalytic domains. The RPTPs are thought to transduce extracellular signals by dephosphorylating tyrosine-phosphorylated intracellular substrates. As such, they are the enzymatic counterparts of the well studied receptor tyrosine kinases. However, little is known about the signaling mechanisms and biological functions of the RPTPs. Recent studies show that the extracellular domain of certain RPTPs can mediate either homophilic or heterophilic interactions and suggest a role in cadherin-mediated cell-cell adhesion, possibly via an action on catenins. This review will focus on the role of RPTPs in cell-cell interaction and the possible biological implications.
- Published
- 1997
- Full Text
- View/download PDF
18. Lysophosphatidic acid: G-protein signalling and cellular responses.
- Author
-
Moolenaar WH, Kranenburg O, Postma FR, and Zondag GC
- Subjects
- Animals, Humans, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Receptors, Lysophosphatidic Acid, GTP-Binding Proteins metabolism, Lysophospholipids metabolism, Receptors, G-Protein-Coupled, Signal Transduction
- Abstract
Lysophosphatidic acid (LPA) is a serum-borne phospholipid that activates a specific G protein coupled receptor to evoke multiple cellular responses. Recent work has identified two cDNAs encoding putative LPA receptors, various LPA-like agonists that act on distinct receptors, and new pathways that link the receptor(s) to such diverse events as Ras signalling, cytoskeletal remodelling and membrane depolarization.
- Published
- 1997
- Full Text
- View/download PDF
19. Lack of association between receptor protein tyrosine phosphatase RPTP mu and cadherins.
- Author
-
Zondag GC, Moolenaar WH, and Gebbink MF
- Subjects
- Animals, Antibodies, Monoclonal, COS Cells chemistry, COS Cells enzymology, Cadherins analysis, Cytoskeletal Proteins analysis, Cytoskeletal Proteins metabolism, Epithelium chemistry, Epithelium enzymology, Gene Expression physiology, Lung cytology, Membrane Proteins genetics, Mink, Precipitin Tests, Protein Tyrosine Phosphatases genetics, Protein Tyrosine Phosphatases immunology, Receptor-Like Protein Tyrosine Phosphatases, Class 2, Receptor-Like Protein Tyrosine Phosphatases, Class 8, Receptors, Cell Surface genetics, beta Catenin, Cadherins metabolism, Membrane Proteins metabolism, Protein Tyrosine Phosphatases metabolism, Receptors, Cell Surface metabolism, Trans-Activators
- Abstract
RPTP mu is a receptor-like protein tyrosine phosphatase that mediates homophilic cell-cell interactions. Surface expression of RPTP mu is restricted to cell-cell contacts and is upregulated with increasing cell density, suggesting a role for RPTP mu in contact-mediated signaling. It was recently reported (Brady-Kalnay, S.M., D.L. Rimm, and N.K. Tonks. 1995. J. Cell Biol. 130:977-986) that RPTP mu binds directly to cadherin/catenin complexes, and thus may regulate the tyrosine phosphorylation of such complexes. Here we report that this concept needs revision. Through reciprocal precipitations using a variety of antibodies against RPTP mu, cadherins, and catenins, we show that RPTP mu does not interact with cadherin/catenin complexes, even when assayed under very mild lysis conditions. We find that the anti-RPTP mu antiserum used by others precipitates cadherins in a nonspecific manner independent of RPTP mu. We conclude that, contrary to previous claims, RPTP mu does not interact with cadherin complexes and thus is unlikely to directly regulate cadherin/catenin function.
- Published
- 1996
- Full Text
- View/download PDF
20. Cell surface expression of receptor protein tyrosine phosphatase RPTP mu is regulated by cell-cell contact.
- Author
-
Gebbink MF, Zondag GC, Koningstein GM, Feiken E, Wubbolts RW, and Moolenaar WH
- Subjects
- 3T3 Cells cytology, 3T3 Cells physiology, Animals, Base Sequence, Cell Count, DNA, Complementary, Gene Expression physiology, Haplorhini, Humans, Membrane Proteins metabolism, Mice, Mink, Molecular Sequence Data, Mutation physiology, Protein Binding physiology, Protein Tyrosine Phosphatases biosynthesis, Protein Tyrosine Phosphatases ultrastructure, Rats, Signal Transduction physiology, Transfection, Up-Regulation physiology, Cell Communication physiology, Protein Tyrosine Phosphatases physiology
- Abstract
RPTP mu is a transmembrane protein tyrosine phosphatase with an adhesion molecule-like ectodomain. It has recently been shown that RPTP mu mediates homophilic interactions when expressed in insect cells. In this study, we have examined how RPTP mu may function as a cell contact receptor in mink lung epithelial cells, which express RPTPmu endogenously, as well as in transfected 3T3 cells. We find that RPTP mu has a relatively short half-life (3-4 hours) and undergoes posttranslational cleavage into two noncovalently associated subunits, with both cleaved and uncleaved molecules being present on the cell surface (roughly at a 1:1 ratio); shedding of the ectodomain subunit is observed in exponentially growing cells. Immunofluorescence analysis reveals that surface expression of RPTPmu is restricted to regions of tight cell-cell contact. RPTPmu surface expression increases significantly with increasing cell density. This density-induced upregulation of RPTP mu is independent of its catalytic activity and is also observed when transcription is driven by a constitutive promoter, indicating that modulation of RPTPmu surface expression occurs posttranscriptionally. Based on our results, we propose the following model of RPTP mu function: In the absence of cell-cell contact, newly synthesized RPTP mu molecules are rapidly cleared from the cell surface. Cell-cell contact causes RPTPmu to be trapped at the surface through homophilic binding, resulting in accumulation of RPTP mu at intercellular contact regions. This contact-induced clustering of RPTPmu may then lead to tyrosine dephosphorylation of intracellular substrates at cell-cell contacts.
- Published
- 1995
- Full Text
- View/download PDF
21. Homophilic interactions mediated by receptor tyrosine phosphatases mu and kappa. A critical role for the novel extracellular MAM domain.
- Author
-
Zondag GC, Koningstein GM, Jiang YP, Sap J, Moolenaar WH, and Gebbink MF
- Subjects
- Animals, Base Sequence, Cell Adhesion, Cell Line, Cloning, Molecular, DNA Primers, Humans, Membrane Proteins genetics, Molecular Sequence Data, Protein Tyrosine Phosphatases genetics, Receptor-Like Protein Tyrosine Phosphatases, Class 2, Receptor-Like Protein Tyrosine Phosphatases, Class 8, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Spodoptera, Membrane Proteins metabolism, Protein Tyrosine Phosphatases metabolism
- Abstract
The receptor-like protein tyrosine phosphatases (RPTP) mu and RPTP kappa have a modular ectodomain consisting of four fibronectin type III-like repeats, a single Ig-like domain, and a newly identified N-terminal MAM domain. The function of the latter module, which comprises about 160 amino acids and is found in diverse transmembrane proteins, is not known. We previously reported that both RPTP mu and RPTP kappa can mediate homophilic cell interactions when expressed in insect cells. Here we show that despite their striking structural similarity, RPTP mu and RPTP kappa fail to interact in a heterophilic manner. To examine the role of the MAM domain in homophilic binding, we expressed a mutant RPTP mu lacking the MAM domain in insect Sf9 cells. Truncated RPTP mu is properly expressed at the cell surface but fails to promote cell-cell adhesion. Homophilic cell adhesion is fully restored in a chimeric RPTP mu molecule containing the MAM domain of RPTP kappa. However, this chimeric RPTP mu does not interact with either RPTP mu or RPTP kappa. These results indicate that the MAM domain of RPTP mu and RPTP kappa is essential for homophilic cell-cell interaction and helps determine the specificity of these interactions.
- Published
- 1995
- Full Text
- View/download PDF
22. Purification and characterization of the cytoplasmic domain of human receptor-like protein tyrosine phosphatase RPTP mu.
- Author
-
Gebbink MF, Verheijen MH, Zondag GC, van Etten I, and Moolenaar WH
- Subjects
- Animals, Baculoviridae genetics, Escherichia coli genetics, Gene Deletion, Humans, Moths, Muramidase metabolism, Mutagenesis, Site-Directed, Myelin Basic Protein metabolism, Phosphotyrosine, Point Mutation, Protein Tyrosine Phosphatases genetics, Protein Tyrosine Phosphatases metabolism, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Structure-Activity Relationship, Substrate Specificity, Tyrosine analogs & derivatives, Tyrosine metabolism, Protein Tyrosine Phosphatases isolation & purification
- Abstract
RPTP mu is a recently described receptor-like protein tyrosine phosphatase (PTP), the ectodomain of which mediates homophilic cell-cell adhesion. The cytoplasmic part contains two homologous PTP-like domains and a juxtamembrane region that is about twice as large as in other receptor-like PTPs. The entire 80-kDa cytoplasmic part of human RPTP mu was expressed in insect Sf9 cells and its enzymatic activity was characterized after purification to electrophoretic homogeneity. In addition, the effects of deletion and point mutations were analyzed following expression in Escherichia coli cells. The purified cytoplasmic part of RPTP mu displays high activity toward tyrosine-phosphorylated, modified lysozyme (Vmax 4500 nmol min-1 mg-1) and myelin basic protein (Vmax 8500 nmol min-1 mg-1) but negligible activity toward tyrosine-phosphorylated angiotensin or the nonapeptide, EDNDpYINASL, that serves as a good substrate for protein tyrosine phosphatase PTP1B. This suggests that RPTP mu and PTP1B have distinct substrate specificities. Catalytic activity is independent of Ca2+ (up to 1 mM) but is strongly inhibited by Zn2+, Mn2+, vanadate, phenylarsenic oxide, and heparin. The first of the two catalytic domains is 5-10 times less active than the expressed catalytic region containing both domains. Mutation of Cys 1095 to Ser in the first catalytic domain abolishes enzymatic activity when analyzed following expression in either E. coli or mammalian COS cells. Deletion of the first 53 amino acids from the juxtamembrane region reduces catalytic activity about 2-fold.
- Published
- 1993
- Full Text
- View/download PDF
23. Cell-cell adhesion mediated by a receptor-like protein tyrosine phosphatase.
- Author
-
Gebbink MF, Zondag GC, Wubbolts RW, Beijersbergen RL, van Etten I, and Moolenaar WH
- Subjects
- Animals, Base Sequence, Calcium metabolism, Catalysis, Cell Line, DNA, Humans, Hydrogen-Ion Concentration, Molecular Sequence Data, Moths, Receptor-Like Protein Tyrosine Phosphatases, Class 2, Receptor-Like Protein Tyrosine Phosphatases, Class 8, Cell Adhesion, Membrane Proteins metabolism, Protein Tyrosine Phosphatases metabolism, Receptors, Cell Surface metabolism
- Abstract
Receptor-like protein tyrosine phosphatases (receptor-PTPs) represent a novel family of transmembrane proteins that are thought to play important roles in cellular regulation. They consist of a cytoplasmic catalytic region, a single transmembrane segment and an extracellular, putative ligand-binding domain, but the nature of their physiological ligands is unknown. We have recently cloned a new receptor-PTP (RPTP mu), the ectodomain of which includes an Ig-like and four fibronectin type III-like domains, suggesting that RPTP mu may be involved in cell-cell or cell-matrix interactions. To test this hypothesis, we expressed RPTP mu in insect Sf9 cells using recombinant baculovirus. We demonstrate that RPTP mu dramatically promotes cell-to-cell adhesion in a homophilic, Ca(2+)-independent manner. No adhesion is observed in Sf9 cells expressing a chimeric RPTP mu molecule containing the extracellular domain of the epidermal growth factor receptor. Furthermore, cells expressing an enzymatically inactive, point-mutated RPTP mu or a truncated form of RPTP mu, lacking the entire catalytic region, show adhesive properties indistinguishable from those of wild-type RPTP mu, indicating that the catalytic domain is not essential for RPTP mu-mediated adhesion. These results assign a physiological role for RPTP mu in signaling cell-cell recognition.
- Published
- 1993
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.