226 results on '"Johannes L. Bos"'
Search Results
2. Drug-repurposing screen on patient-derived organoids identifies therapy-induced vulnerability in KRAS-mutant colon cancer
- Author
-
Sander Mertens, Maarten A. Huismans, Carla S. Verissimo, Bas Ponsioen, Rene Overmeer, Natalie Proost, Olaf van Tellingen, Marieke van de Ven, Harry Begthel, Sylvia F. Boj, Hans Clevers, Jeanine M.L. Roodhart, Johannes L. Bos, and Hugo J.G. Snippert
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
3. CRISPR-induced RASGAP deficiencies in colorectal cancer organoids reveal that only loss of NF1 promotes resistance to EGFR inhibition
- Author
-
Suraya Elfrink, Hugo J. Snippert, Nizar Hami, Johannes L. Bos, Jasmin B. Post, and Alexander E E Mertens
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,RASGAP ,Colorectal cancer ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,colorectal cancer ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Organoid ,medicine ,CRISPR ,neoplasms ,Mechanism (biology) ,business.industry ,Egfr inhibition ,medicine.disease ,cancer progression ,digestive system diseases ,anti-EGFR therapy resistance ,030104 developmental biology ,Oncology ,NF1 ,030220 oncology & carcinogenesis ,Cancer research ,Biomarker (medicine) ,KRAS ,business ,Research Paper - Abstract
Contains fulltext : 215760.pdf (Publisher’s version ) (Open Access) Anti-EGFR therapy is used to treat metastatic colorectal cancer (CRC) patients, for which initial response rates of 10-20% have been achieved. Although the presence of HER2 amplifications and oncogenic mutations in KRAS, NRAS, and BRAF are associated with EGFR-targeted therapy resistance, for a large population of CRC patients the underlying mechanism of RAS-MEK-ERK hyperactivation is not clear. Loss-of-function mutations in RASGAPs are often speculated in literature to promote CRC growth as being negative regulators of RAS, but direct experimental evidence is lacking. We generated a CRISPR-mediated knock out panel of all RASGAPs in patient-derived CRC organoids and found that only loss of NF1, but no other RASGAPs e.g. RASA1, results in enhanced RAS-ERK signal amplification and improved tolerance towards limited EGF stimulation. Our data suggests that NF1-deficient CRCs are likely not responsive to anti-EGFR monotherapy and can potentially function as a biomarker for CRC progression.
- Published
- 2019
- Full Text
- View/download PDF
4. Drug Repurposing Screen on Patient-Derived Organoids Identifies New Therapeutic Drug Combination Against KRAS Mutant Colon Cancer
- Author
-
Hugo J. Snippert, Marieke van de Ven, Natalie Proost, René M. Overmeer, Carla S Verissimo, Hans Clevers, Johannes L. Bos, Jeanine M.L. Roodhart, Olaf van Tellingen, Harry Begthel, Jeanine Roodhart, Maarten A. Huismans, Bas Ponsioen, Maarten Huismans, Sylvia F. Boj, and Sander Mertens
- Subjects
Drug ,Colorectal cancer ,business.industry ,media_common.quotation_subject ,Cell cycle ,medicine.disease ,medicine.disease_cause ,Vinorelbine ,Drug repositioning ,Cell killing ,Tolerability ,medicine ,Cancer research ,KRAS ,business ,media_common ,medicine.drug - Abstract
Patient-derived organoids (PDO) are widely heralded as a drug-screening platform to develop new anti-cancer therapies. Here we use a drug-repurposing library to screen PDOs of colorectal cancer (CRC) to identify drugs that enhance the efficacy of combinatorial targeting of EGFR and MEK in KRAS mutant CRCs. Although this combination achieves significant inhibition of downstream signaling of mutant KRAS, preclinical models revealed its failure to induce robust cell killing. We developed a microscopy-based screen to score drug-induced cytotoxicity in PDOs and tested 414 putative anti-cancer drugs in combination with EGFR/MEK inhibition. A majority of validated hits were microtubule targeting agents that are commonly used in clinical oncology, like taxanes and vinca-alkaloids. One of these drugs, vinorelbine, was consistently effective across a panel of >20 different CRC PDOs. Unlike vinorelbine alone, its combination with EGFR/MEK inhibition induces apoptosis at all stages of the cell cycle, presumably by disrupting the balance between pro- and anti-apoptotic effectors. In mice, the triple combination showed tolerability and superior antitumor activity at clinically relevant doses and scheduling, setting the basis for a clinical trial to treat patients with metastatic RAS mutant CRC.
- Published
- 2021
- Full Text
- View/download PDF
5. Quantifying single-cell ERK dynamics in colorectal cancer organoids reveals EGFR as an amplifier of oncogenic MAPK pathway signalling
- Author
-
Livio Trusolino, Francesco Sassi, Sander Mertens, Benjamin Cappe, Ingrid Verlaan-Klink, Ravian L. van Ineveld, Bas Ponsioen, Sylvia F. Boj, Simone Kersten, Julian R. Buissant des Amorie, Dimitrios Laskaris, Rob G. J. Vries, Franck B. Riquet, Andrea Bertotti, Jasmin B. Post, François Sipieter, Johannes L. Bos, Peter Vandenabeele, Hugo J. Snippert, Holger Rehmann, University Medical Center [Utrecht], Department of Biomedical Molecular Biology [Ghent], Universiteit Gent = Ghent University [Belgium] (UGENT), Institut Jacques Monod (IJM (UMR_7592)), and Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)
- Subjects
MAPK/ERK pathway ,endocrine system diseases ,cell-to-cell heterogeneity ,pan-HER inhibition ,[SDV]Life Sciences [q-bio] ,Mitogen-activated protein kinase kinase ,medicine.disease_cause ,0302 clinical medicine ,Epidermal growth factor receptor ,oncogenic signaling ,EGFR inhibitors ,0303 health sciences ,Tumor ,biology ,Kinase ,Chemistry ,3. Good health ,Cell biology ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Organoids ,030220 oncology & carcinogenesis ,KRAS ,Signal transduction ,Single-Cell Analysis ,Colorectal Neoplasms ,patient-derived organoids ,Proto-Oncogene Proteins B-raf ,MAP Kinase Signaling System ,EGFR ,FRET biosensors ,Article ,Cell Line ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Humans ,Protein kinase A ,neoplasms ,Protein Kinase Inhibitors ,030304 developmental biology ,Mitogen-Activated Protein Kinase Kinases ,Neoplastic ,Cell Biology ,ERK oscillations ,Colorectal cancer ,digestive system diseases ,Gene Expression Regulation ,Mutation ,biology.protein - Abstract
Direct targeting of the downstream mitogen-activated protein kinase (MAPK) pathway to suppress extracellular-regulated kinase (ERK) activation in KRAS and BRAF mutant colorectal cancer (CRC) has proven clinically unsuccessful, but promising results have been obtained with combination therapies including epidermal growth factor receptor (EGFR) inhibition. To elucidate the interplay between EGF signalling and ERK activation in tumours, we used patient-derived organoids (PDOs) from KRAS and BRAF mutant CRCs. PDOs resemble in vivo tumours, model treatment response and are compatible with live-cell microscopy. We established real-time, quantitative drug response assessment in PDOs with single-cell resolution, using our improved fluorescence resonance energy transfer (FRET)-based ERK biosensor EKAREN5. We show that oncogene-driven signalling is strikingly limited without EGFR activity and insufficient to sustain full proliferative potential. In PDOs and in vivo, upstream EGFR activity rigorously amplifies signal transduction efficiency in KRAS or BRAF mutant MAPK pathways. Our data provide a mechanistic understanding of the effectivity of EGFR inhibitors within combination therapies against KRAS and BRAF mutant CRC. Ponsioen et al. use a FRET‐based ERK biosensor EKAREN5 in patient‐derived organoids to show that EGFR activity amplifies signal transduction efficiency in KRAS or BRAF mutant MAPK pathways.
- Published
- 2020
- Full Text
- View/download PDF
6. Multiple Rap1 effectors control Epac1-mediated tightening of endothelial junctions
- Author
-
Marjolein J. Vliem, Willem-Jan Pannekoek, and Johannes L. Bos
- Subjects
endocrine system ,junctional actin ,endothelial barrier ,Kinesins ,FGD5 ,Biology ,Myosins ,Circumferential Actin Cables ,Biochemistry ,Tight Junctions ,03 medical and health sciences ,0302 clinical medicine ,Endothelial barrier ,Guanine Nucleotide Exchange Factors ,Humans ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Tension (physics) ,Effector ,Endothelial Cells ,rap1 GTP-Binding Proteins ,Cell Biology ,AF6 ,humanities ,Cell biology ,Radil, Rap1 ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,030220 oncology & carcinogenesis ,Research Paper/Report ,Rap1 ,cytoskeletal tension ,Radial stress ,Research Article - Abstract
Epac1 and Rap1 mediate cAMP-induced tightening of endothelial junctions. We have previously found that one of the mechanisms is the inhibition of Rho-mediated tension in radial stress fibers by recruiting the RhoGAP ArhGAP29 in a complex containing the Rap1 effectors Rasip1 and Radil. However, other mechanisms have been proposed as well, most notably the induction of tension in circumferential actin cables by Cdc42 and its GEF FGD5. Here, we have investigated how Rap1 controls FGD5/Cdc42 and how this interconnects with Radil/Rasip1/ArhGAP29. Using endothelial barrier measurements, we show that Rho inhibition is not sufficient to explain the barrier stimulating effect of Rap1. Indeed, Cdc42-mediated tension is induced at cell-cell contacts upon Rap1 activation and this is required for endothelial barrier function. Depletion of potential Rap1 effectors identifies AF6 to mediate Rap1 enhanced tension and concomitant Rho-independent barrier function. When overexpressed in HEK293T cells, AF6 is found in a complex with FGD5 and Radil. From these results we conclude that Rap1 utilizes multiple pathways to control tightening of endothelial junctions, possibly through a multiprotein effector complex, in which AF6 functions to induce tension in circumferential actin cables.
- Published
- 2020
- Full Text
- View/download PDF
7. P119 An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity
- Author
-
Trudy G. N. Jonges, LM van Wijk, Mpg Vreeswijk, Hans Clevers, Kadi Lõhmussaar, MJ van Roosmalen, M van de Ven, Johannes L. Bos, A van Oudenaarden, Oded Kopper, Nizar Hami, SA Revilla, Vwh Ho, Harry Begthel, Hjg Snippert, Wigard P. Kloosterman, J Espejo Valle-Inclan, Petronella O. Witteveen, P. J. Van Diest, Katja N. Gaarenstroom, Benjamin G. Neel, Harry Vrieling, Ronald P. Zweemer, Lennart Kester, Jeroen Korving, Tjalling Bosse, Natalie Proost, R Theeuwsen, Bas Ponsioen, CJ de Witte, and Anjali Vanita Balgobind
- Subjects
Oncology ,medicine.medical_specialty ,Competing interests ,Tumour heterogeneity ,business.industry ,Late stage ,Cancer ,Biology ,medicine.disease ,Internal medicine ,medicine ,Organoid ,Recurrent disease ,Personalized medicine ,Ovarian cancer ,business - Abstract
Introduction/Background Ovarian cancer (OC) is a heterogeneous disease usually diagnosed at a late stage. Experimental in vitro models that faithfully capture the hallmarks and tumour heterogeneity of OC are limited and hard to establish. Methodology We present a protocol that enables efficient derivation and long-term expansion of OC organoids. Results Utilizing this protocol, we have established 56 organoid lines from 32 patients, representing all main subtypes of OC. OC organoids recapitulate histological and genomic features of the pertinent lesion from which they were derived, illustrating intra- and interpatient heterogeneity, and can be genetically modified. We show that OC organoids can be used for drug-screening assays and capture different tumour subtype responses to the gold standard platinum-based chemotherapy, including acquisition of chemoresistance in recurrent disease. Finally, OC organoids can be xenografted, enabling in vivo drug-sensitivity assays. Conclusion Taken together, this demonstrates their potential application for research and personalized medicine. Disclosure No competing interests. Applicable funding sources: EIF | Stand Up To Cancer (SU2C) [Clevers, Kopper, Balgobind, Begthel] KWF Kankerbestrijding (Dutch Cancer Society) - UU2015-7743 [de Witte, van Roosmalen, Witteveen, Zweemer, Kloosterman] Gieskes Strijbis Foundation (1816199) [Clevers, Witteveen, Kloosterman, Zweemer, Lohmussaar, Espejo Valle-Inclan, Hami, Bos, Snippert] Published in Nature Medicine. April 2019. dx.doi.org/10.1038/s41591-019-0422-6.
- Published
- 2019
- Full Text
- View/download PDF
8. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity
- Author
-
Jose Espejo Valle-Inclan, Victor W.H. Ho, Nizar Hami, Markus J. van Roosmalen, Benjamin G. Neel, Kadi Lõhmussaar, Anjali Vanita Balgobind, Lennart Kester, Ronald P. Zweemer, Harry Begthel, Harry Vrieling, Jeroen Korving, Tjalling Bosse, Chris J. de Witte, Maaike P.G. Vreeswijk, Oded Kopper, Wigard P. Kloosterman, Natalie Proost, Alexander van Oudenaarden, Marieke van de Ven, Paul J. van Diest, Rebecca Theeuwsen, Bas Ponsioen, Johannes L. Bos, Hugo J. Snippert, Katja N. Gaarenstroom, Lise M van Wijk, Trudy N. Jonges, Hans Clevers, Petronella O. Witteveen, Sonia Aristín Revilla, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
0301 basic medicine ,Adult ,Disease ,Mice, SCID ,Biology ,Drug Screening Assays ,SCID ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,medicine ,Recurrent disease ,Organoid ,Journal Article ,Animals ,Humans ,Precision Medicine ,Aged ,Tumor ,business.industry ,Ovarian Neoplasms/drug therapy ,General Medicine ,Antitumor ,Genomics ,Middle Aged ,medicine.disease ,Precision medicine ,Tumor Subtype ,Organoids/pathology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Heterografts ,Female ,Personalized medicine ,Drug Screening Assays, Antitumor ,business ,Ovarian cancer - Abstract
Ovarian cancer (OC) is a heterogeneous disease usually diagnosed at a late stage. Experimental in vitro models that faithfully capture the hallmarks and tumor heterogeneity of OC are limited and hard to establish. We present a protocol that enables efficient derivation and long-term expansion of OC organoids. Utilizing this protocol, we have established 56 organoid lines from 32 patients, representing all main subtypes of OC. OC organoids recapitulate histological and genomic features of the pertinent lesion from which they were derived, illustrating intra- and interpatient heterogeneity, and can be genetically modified. We show that OC organoids can be used for drug-screening assays and capture different tumor subtype responses to the gold standard platinum-based chemotherapy, including acquisition of chemoresistance in recurrent disease. Finally, OC organoids can be xenografted, enabling in vivo drug-sensitivity assays. Taken together, this demonstrates their potential application for research and personalized medicine. A biobank of ovarian cancer organoids recapitulates the histopathological and molecular hallmarks of patient tumors and provides a resource for preclinical research.
- Published
- 2019
9. The Phosphatase PTPL1 Is Required for PTEN-Mediated Regulation of Apical Membrane Size
- Author
-
Rosendahl Huber Akm, Susan Zwakenberg, Johannes L. Bos, Bruurs Ljm, Anneke Post, Fried J. T. Zwartkruis, and van der Net Mc
- Subjects
0301 basic medicine ,Male ,PTEN ,Phosphatase ,Protein Tyrosine Phosphatase, Non-Receptor Type 13 ,Protein tyrosine phosphatase ,03 medical and health sciences ,Gene Knockout Techniques ,Mice ,Cell Line, Tumor ,Neoplasms ,Cell cortex ,Cell polarity ,medicine ,Animals ,Humans ,Spotlight ,Molecular Biology ,biology ,Microvilli ,HEK 293 cells ,Cell Membrane ,PTEN Phosphohydrolase ,Cell Polarity ,Epithelial Cells ,Cell Biology ,Apical membrane ,Epithelium ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,PTPL1 ,biology.protein ,CRISPR-Cas Systems ,Research Article - Abstract
PTEN is a tumor suppressor that is frequently lost in epithelial malignancies. A part of the tumor-suppressive properties of PTEN is attributed to its function in cell polarization and consequently its role in maintaining epithelial tissue integrity. However, surprisingly little is known about the function and regulation of PTEN during epithelial cell polarization. We used clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9-mediated gene disruption to delete PTEN in intestinal epithelial Ls174T:W4 cells, which upon differentiation form a microvillus-covered apical membrane (brush border) on a part of the cell cortex, independent of cell-cell junctions. We show that loss of PTEN results in the formation of a larger brush border that, in a fraction of the cells, even spans the entire plasma membrane, revealing that PTEN functions in the regulation of apical membrane size. Depletion of the phosphatase PTPL1 resulted in a similar defect. PTPL1 interacts with PTEN, and this interaction is necessary for apical membrane enrichment of PTEN. Importantly, phosphatase activity of PTPL1 is not required, indicating that PTPL1 functions as an anchor protein in this process. Our work thus demonstrates a novel function for PTEN during cell polarization in controlling apical membrane size and identifies PTPL1 as a critical apical membrane anchor for PTEN in this process.
- Published
- 2018
10. ATP8B1-mediated spatial organization of Cdc42 signaling maintains singularity during enterocyte polarization
- Author
-
George Posthuma, Harry Begthel, Stan F.J. van de Graaf, Johannes L. Bos, A.S. Knisely, Coen C. Paulusma, Lisa Donker, Susan Zwakenberg, Lucas J. M. Bruurs, Fried J. T. Zwartkruis, Amsterdam Gastroenterology Endocrinology Metabolism, Gastroenterology and Hepatology, Amsterdam institute for Infection and Immunity, and Tytgat Institute for Liver and Intestinal Research
- Subjects
Endosome ,Enterocyte ,Membrane lipids ,macromolecular substances ,CDC42 ,Biology ,Cell Line ,Membrane Lipids ,Mice ,Report ,Cell polarity ,Journal Article ,medicine ,Animals ,Humans ,Phospholipid Transfer Proteins ,cdc42 GTP-Binding Protein ,Research Articles ,Adenosine Triphosphatases ,Cell Polarity ,Cell Biology ,Flippase ,Apical membrane ,Cell biology ,Enterocytes ,medicine.anatomical_structure ,Cdc42 GTP-Binding Protein ,Signal Transduction - Abstract
The disease-associated phospholipid flippase ATP8B1 decreases Cdc42 mobility at the apical membrane to ensure the formation of a single apical domain and to maintain healthy lumen architecture., During yeast cell polarization localization of the small GTPase, cell division control protein 42 homologue (Cdc42) is clustered to ensure the formation of a single bud. Here we show that the disease-associated flippase ATPase class I type 8b member 1 (ATP8B1) enables Cdc42 clustering during enterocyte polarization. Loss of this regulation results in increased apical membrane size with scattered apical recycling endosomes and permits the formation of more than one apical domain, resembling the singularity defect observed in yeast. Mechanistically, we show that to become apically clustered, Cdc42 requires the interaction between its polybasic region and negatively charged membrane lipids provided by ATP8B1. Disturbing this interaction, either by ATP8B1 depletion or by introduction of a Cdc42 mutant defective in lipid binding, increases Cdc42 mobility and results in apical membrane enlargement. Re-establishing Cdc42 clustering, by tethering it to the apical membrane or lowering its diffusion, restores normal apical membrane size in ATP8B1-depleted cells. We therefore conclude that singularity regulation by Cdc42 is conserved between yeast and human and that this regulation is required to maintain healthy tissue architecture.
- Published
- 2015
- Full Text
- View/download PDF
11. Rap1 Spatially Controls ArhGAP29 To Inhibit Rho Signaling during Endothelial Barrier Regulation
- Author
-
Bas Ponsioen, Willem-Jan Pannekoek, Marjolein J. Vliem, Anneke Post, and Johannes L. Bos
- Subjects
rho GTP-Binding Proteins ,endocrine system ,GTPase-activating protein ,GTPase ,Biology ,Human Umbilical Vein Endothelial Cells ,Guanine Nucleotide Exchange Factors ,Humans ,Small GTPase ,Molecular Biology ,Cells, Cultured ,Microscopy, Confocal ,GTPase-Activating Proteins ,Intracellular Signaling Peptides and Proteins ,rap1 GTP-Binding Proteins ,Articles ,Cell Biology ,Actin cytoskeleton ,Cell biology ,Luminescent Proteins ,Protein Transport ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,Intercellular Junctions ,RNA Interference ,Rap1 ,MDia1 ,Guanine nucleotide exchange factor ,Signal transduction ,Carrier Proteins ,Protein Binding ,Signal Transduction - Abstract
The small GTPase Rap1 controls the actin cytoskeleton by regulating Rho GTPase signaling. We recently established that the Rap1 effectors Radil and Rasip1, together with the Rho GTPase activating protein ArhGAP29, mediate Rap1-induced inhibition of Rho signaling in the processes of epithelial cell spreading and endothelial barrier function. Here, we show that Rap1 induces the independent translocations of Rasip1 and a Radil-ArhGAP29 complex to the plasma membrane. This results in the formation of a multimeric protein complex required for Rap1-induced inhibition of Rho signaling and increased endothelial barrier function. Together with the previously reported spatiotemporal control of the Rap guanine nucleotide exchange factor Epac1, these findings elucidate a signaling pathway for spatiotemporal control of Rho signaling that operates by successive protein translocations to and complex formation at the plasma membrane.
- Published
- 2015
- Full Text
- View/download PDF
12. A Tuba/Cdc42/Par6A complex is required to ensure singularity in apical domain formation during enterocyte polarization
- Author
-
Johannes L. Bos, Mirjam C. van der Net, Susan Zwakenberg, Lucas J. M. Bruurs, and Fried J. T. Zwartkruis
- Subjects
0301 basic medicine ,Hydrolases ,Cell Membranes ,lcsh:Medicine ,CDC42 ,Biochemistry ,Epithelium ,Signaling Molecules ,Gene Knockout Techniques ,Database and Informatics Methods ,0302 clinical medicine ,Cell Signaling ,Animal Cells ,Cell polarity ,Medicine and Health Sciences ,Guanine Nucleotide Exchange Factors ,Small GTPase ,cdc42 GTP-Binding Protein ,lcsh:Science ,Multidisciplinary ,Microvilli ,Chemistry ,Cell Polarity ,Signal transducing adaptor protein ,Precipitation Techniques ,Enzymes ,Cell biology ,medicine.anatomical_structure ,Guanine nucleotide exchange factor ,Cellular Structures and Organelles ,Cellular Types ,Anatomy ,Signal transduction ,Sequence Analysis ,Signal Transduction ,Research Article ,Cell Physiology ,Bioinformatics ,Enterocyte ,Research and Analysis Methods ,Cell Line ,03 medical and health sciences ,Sequence Motif Analysis ,medicine ,Humans ,Immunoprecipitation ,Protein Structure, Quaternary ,GTPase signaling ,Adaptor Proteins, Signal Transducing ,lcsh:R ,Biology and Life Sciences ,Proteins ,Epithelial Cells ,Cell Biology ,Apical membrane ,Co-Immunoprecipitation ,Cytoskeletal Proteins ,Guanosine Triphosphatase ,Enterocytes ,Biological Tissue ,030104 developmental biology ,Enzymology ,lcsh:Q ,030217 neurology & neurosurgery - Abstract
Apico-basal polarity establishment is a seminal process in tissue morphogenesis. To function properly it is often imperative that epithelial cells limit apical membrane formation to a single domain. We previously demonstrated that signaling by the small GTPase Cdc42, together with its guanine nucleotide exchange factor (GEF) Tuba, is required to prevent the formation of multiple apical domains in polarized Ls174T:W4 cells, a single cell model for enterocyte polarization. To further chart the molecular signaling mechanisms that safeguard singularity during enterocyte polarization we generated knockout cells for the Cdc42 effector protein Par6A. Par6A loss results in the formation of multiple apical domains, similar to loss of Cdc42. In Par6A knockout cells, we find that active Cdc42 is more mobile at the apical membrane compared to control cells and that wild type Cdc42 is more diffusely localized throughout the cell, indicating that Par6A is required to restrict Cdc42 signaling. Par6A, Cdc42 and its GEF Tuba bind in a co-immunoprecipitation experiment and they partially colocalize at the apical membrane in polarized Ls174T:W4 cells, suggesting the formation of a trimeric complex. Indeed, in a rescue experiment using Par6A mutants, we show that the ability to establish this trimeric complex correlates with the ability to restore singularity in Par6A knockout cells. Together, these experiments therefore indicate that a Tuba/Cdc42/Par6A complex is required to ensure the formation of a single apical domain during enterocyte polarization.
- Published
- 2018
13. From Ras to Rap and Back, a Journey of 35 Years
- Author
-
Johannes L. Bos
- Subjects
0301 basic medicine ,Mutation ,Oncogene ,Retrospective ,Oncogenes ,Biology ,medicine.disease_cause ,Actin cytoskeleton ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Closest relatives ,03 medical and health sciences ,Signaling network ,030104 developmental biology ,rap GTP-Binding Proteins ,medicine ,Cell Adhesion ,ras Proteins ,Animals ,Humans ,Signal transduction ,Cell adhesion ,Early analysis ,Signal Transduction - Abstract
Our laboratory has studied Ras and Ras-like proteins since the discovery of the Ras oncogene 35 years ago. In this review, I will give an account of what we have done in these 35 years and indicate the main papers that have guided our research. Our efforts started with the early analysis of mutant Ras in human tumors followed by deciphering of the role of Ras in signal transduction pathways. In an attempt to interfere in Ras signaling we turned to Rap proteins. These proteins are the closest relatives of Ras and were initially identified as Ras antagonists. However, our studies revealed that the Rap signaling network primarily is involved in spatiotemporal control of cell adhesion, in part through regulation of the actin cytoskeleton. More recently we returned to Ras, trying to interfere in Ras signaling by combinatorial drug testing using the organoid technology.
- Published
- 2017
14. A Two-Tiered Mechanism Enables Localized Cdc42 Signaling during Enterocyte Polarization
- Author
-
Susan Zwakenberg, Johannes L. Bos, Mirjam C. van der Net, Lucas J. M. Bruurs, and Fried J. T. Zwartkruis
- Subjects
0301 basic medicine ,Cell signaling ,GTP' ,Enterocyte ,macromolecular substances ,CDC42 ,Biology ,Cell Line ,03 medical and health sciences ,Tubulin ,Cell polarity ,medicine ,Humans ,rho-Specific Guanine Nucleotide Dissociation Inhibitors ,Small GTPase ,Cdc42 ,cdc42 GTP-Binding Protein ,Molecular Biology ,GTPase signaling ,Microvilli ,Polarized epithelia ,Cell Membrane ,Cell Biology ,Apical membrane ,Cell biology ,Enterocytes ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,Guanine nucleotide exchange factor ,biological phenomena, cell phenomena, and immunity ,Protein Binding ,Signal Transduction ,Research Article - Abstract
Signaling by the small GTPase Cdc42 governs a diverse set of cellular processes that contribute to tissue morphogenesis. Since these processes often require highly localized signaling, Cdc42 activity must be clustered in order to prevent ectopic signaling. During cell polarization, apical Cdc42 signaling directs the positioning of the nascent apical membrane. However, the molecular mechanisms that drive Cdc42 clustering during polarity establishment are largely unknown. Here, we demonstrate that during cell polarization localized Cdc42 signaling is enabled via activity-dependent control of Cdc42 mobility. By performing photoconversion experiments, we show that inactive Cdc42-GDP is 30-fold more mobile than active Cdc42-GTP. This switch in apical mobility originates from a dual mechanism involving RhoGDI-mediated membrane dissociation of Cdc42-GDP and Tuba-mediated immobilization of Cdc42-GTP. Interference with either mechanism affects Cdc42 clustering and as a consequence impairs Cdc42-mediated apical membrane clustering. We therefore identify a molecular network, comprised of Cdc42, the guanine nucleotide exchange factor (GEF) Tuba, and RhoGDI, that enables differential diffusion of inactive and active Cdc42 and is required to establish localized Cdc42 signaling during enterocyte polarization.
- Published
- 2017
- Full Text
- View/download PDF
15. Prehabilitation worth it? A theoretical algorithmic for the cost-effectiveness of a multimodal prehabilitation program for complex abdominal surgery
- Author
-
L. van Wijk, Johannes L. Bos, and Joost M. Klaase
- Subjects
medicine.medical_specialty ,Hepatology ,business.industry ,Cost effectiveness ,Prehabilitation ,Gastroenterology ,Physical therapy ,Medicine ,business ,Abdominal surgery - Published
- 2020
- Full Text
- View/download PDF
16. Exchange protein activated by cAMP 1(Epac1)‐deficient mice develop β‐cell dysfunction and metabolic syndrome
- Author
-
Xinmei Zhang, Stephen S.M. Chung, Andrew C. P. Tai, Sookja K. Chung, Aimin Xu, Alan K. L. Kai, Angela K. W. Lai, Patrick Ka Kit Yeung, Karen S. Lam, Jian Wang, Sidney Tam, Amy K. M. Lam, Paul M. Vanhoutte, Ying Xian Chen, and Johannes L. Bos
- Subjects
Blood Glucose ,endocrine system ,medicine.medical_specialty ,Biochemistry ,Diabetes Mellitus, Experimental ,Mice ,chemistry.chemical_compound ,Insulin resistance ,Insulin-Secreting Cells ,Internal medicine ,Genetics ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Cyclic adenosine monophosphate ,Obesity ,Molecular Biology ,Embryonic Stem Cells ,Metabolic Syndrome ,Mice, Knockout ,geography ,geography.geographical_feature_category ,biology ,medicine.disease ,Islet ,Streptozotocin ,Dietary Fats ,Endocrinology ,chemistry ,Apoptosis ,biology.protein ,GLUT2 ,Metabolic syndrome ,Insulitis ,Biotechnology ,medicine.drug - Abstract
Previously, exchange protein directly activated by cAMP 2 (Epac2) and PKA were known to play a role in glucose-stimulated insulin secretion (GSIS) by pancreatic β cells. The present study shows that Epac1 mRNA is also expressed by β cells. Therefore, we generated mice and embryonic stem (ES) cells with deletion of the Epac1 gene to define its role in β-cell biology and metabolism. The homozygous Epac1-knockout (Epac1(-/-)) mice developed impaired glucose tolerance and GSIS with deranged islet cytoarchitecture, which was confirmed by isolated islets from adult Epac1(-/-) mice. Moreover, Epac1(-/-) mice developed more severe hyperglycemia with increased β-cell apoptosis and insulitis after multiple low-dose streptozotocin (MLDS; 40 mg/kg) treatment than Epac1(+/+) mice. Interestingly, Epac1(-/-) mice also showed metabolic defects, including increased respiratory exchange ratio (RER) and plasma triglyceride (TG), and more severe diet-induced obesity with insulin resistance, which may contributed to β-cell dysfunction. However, islets differentiated from Epac1(-/-) ES cells showed insulin secretion defect, reduced Glut2 and PDX-1 expression, and abolished GLP-1-stimulated PCNA induction, suggesting a role of Epac1 in β-cell function. The current study provides in vitro and in vivo evidence that Epac1 has an important role in GSIS of β cells and phenotype resembling metabolic syndrome.
- Published
- 2013
- Full Text
- View/download PDF
17. Rasip1 mediates Rap1 regulation of Rho in endothelial barrier function through ArhGAP29
- Author
-
Sarah H. Ross, Anneke Post, Willem-Jan Pannekoek, Patricia M. Brouwer, Johannes L. Bos, and Ingrid Verlaan
- Subjects
endocrine system ,Multidisciplinary ,Stress fiber ,GTPase-activating protein ,GTPase-Activating Proteins ,Intracellular Signaling Peptides and Proteins ,rap1 GTP-Binding Proteins ,Signal transducing adaptor protein ,Context (language use) ,Biological Sciences ,Biology ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Humans ,Rap1 ,Small GTPase ,Endothelium, Vascular ,Signal transduction ,Cells, Cultured ,Actin ,Protein Binding ,Signal Transduction - Abstract
Rap1 is a small GTPase regulating cell–cell adhesion, cell–matrix adhesion, and actin rearrangements, all processes dynamically coordinated during cell spreading and endothelial barrier function. Here, we identify the adaptor protein ras-interacting protein 1 (Rasip1) as a Rap1-effector involved in cell spreading and endothelial barrier function. Using Förster resonance energy transfer, we show that Rasip1 interacts with active Rap1 in a cellular context. Rasip1 mediates Rap1-induced cell spreading through its interaction partner Rho GTPase-activating protein 29 (ArhGAP29), a GTPase activating protein for Rho proteins. Accordingly, the Rap1–Rasip1 complex induces cell spreading by inhibiting Rho signaling. The Rasip1–ArhGAP29 pathway also functions in Rap1-mediated regulation of endothelial junctions, which controls endothelial barrier function. In this process, Rasip1 cooperates with its close relative ras-association and dilute domain-containing protein (Radil) to inhibit Rho-mediated stress fiber formation and induces junctional tightening. These results reveal an effector pathway for Rap1 in the modulation of Rho signaling and actin dynamics, through which Rap1 modulates endothelial barrier function.
- Published
- 2013
- Full Text
- View/download PDF
18. Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening
- Author
-
Hugo J. Snippert, Marc van de Wetering, Johannes L. Bos, Marieke van der Ven, René M. Overmeer, Jarno Drost, David A. Egan, Bastiaan van Gerwen, Hans Clevers, Carla S Verissimo, René Bernards, Sander Mertens, Ingrid Verlaan-Klink, Bas Ponsioen, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
0301 basic medicine ,Colorectal cancer ,QH301-705.5 ,medicine.medical_treatment ,Science ,Drug Evaluation, Preclinical ,Antineoplastic Agents ,colorectal cancer ,Biology ,Bioinformatics ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Targeted therapy ,03 medical and health sciences ,Genome editing ,Organoid ,medicine ,Journal Article ,KRAS ,Humans ,Clustered Regularly Interspaced Short Palindromic Repeats ,Biology (General) ,organoids ,Cancer Biology ,Recombination, Genetic ,General Immunology and Microbiology ,General Neuroscience ,Cancer ,General Medicine ,medicine.disease ,targeted therapy ,030104 developmental biology ,Developmental Biology and Stem Cells ,Cancer cell ,ras Proteins ,Medicine ,Mutant Proteins ,Stem cell ,Colorectal Neoplasms ,Research Article ,Human - Abstract
Colorectal cancer (CRC) organoids can be derived from almost all CRC patients and therefore capture the genetic diversity of this disease. We assembled a panel of CRC organoids carrying either wild-type or mutant RAS, as well as normal organoids and tumor organoids with a CRISPR-introduced oncogenic KRAS mutation. Using this panel, we evaluated RAS pathway inhibitors and drug combinations that are currently in clinical trial for RAS mutant cancers. Presence of mutant RAS correlated strongly with resistance to these targeted therapies. This was observed in tumorigenic as well as in normal organoids. Moreover, dual inhibition of the EGFR-MEK-ERK pathway in RAS mutant organoids induced a transient cell-cycle arrest rather than cell death. In vivo drug response of xenotransplanted RAS mutant organoids confirmed this growth arrest upon pan-HER/MEK combination therapy. Altogether, our studies demonstrate the potential of patient-derived CRC organoid libraries in evaluating inhibitors and drug combinations in a preclinical setting. DOI: http://dx.doi.org/10.7554/eLife.18489.001, eLife digest Recent technical advances mean that miniature replicas of many tissues can be grown in the laboratory. These so-called organoids provide scientists with model systems that are not as limited as simple, two-dimensional sheets of cells growing in a petri dish, and less labor and resource intensive than studies using laboratory animals. In particular, organoids grown from tumor cells from cancer patients have been suggested as having numerous advantages over both laboratory-grown cancer cells and mice when it comes to testing potential new anticancer drugs. Mutations in a gene called KRAS are common in many types of cancer including colon cancer. Tumors with these mutations are difficult to treat and so far virtually all attempts to generate compounds that selectively interfere with the KRAS protein encoded by the mutant gene have failed. Instead, drugs that indirectly inhibit this protein’s effects by targeting other proteins in the same signaling pathway are currently being tested on patients. However, there is still a need for better ways to pre-test whether these drugs will be effective in humans without having to expose the patient to side effects or an ineffective drug. Now, Verissimo, Overmeer, Ponsioen et al. have tested clinically-used KRAS pathway inhibitors and drug combinations against normal colon organoids and colon cancer organoids derived from patients with colon cancer. Gene editing techniques were used to introduce KRAS mutations into some of the normal organoids grown from healthy tissue, and into cancer organoids grown from tumors that had a normal copy of the KRAS gene. In all cases, only those organoids with mutant forms of the KRAS gene were resistant to the treatments. Furthermore, when organoids with the KRAS mutation were treated with some combination therapies that are currently being tested in clinical trials, the tumors stopped growing but the tumor cells failed to die. Similar drug treatments on mice carrying human colon cancer organoids confirmed these results, which is in line with previous studies where tumor tissue from human patients was transplanted into mice. These findings show that collections of tumor organoids from multiple patients could help researchers to quickly identify and optimize targeted anticancer therapies before they are incorporated into clinical trials. In the future, clinical studies are needed to verify how accurately the testing of cancer drugs on organoids predicts whether the drug will or will not work in patients. DOI: http://dx.doi.org/10.7554/eLife.18489.002
- Published
- 2016
19. Author response: Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening
- Author
-
Ingrid Verlaan-Klink, Sander Mertens, Bastiaan van Gerwen, René M. Overmeer, Marc van de Wetering, Marieke van der Ven, Johannes L. Bos, Hugo J. Snippert, Jarno Drost, David A. Egan, Bas Ponsioen, Carla S Verissimo, Hans Clevers, and René Bernards
- Subjects
Drug ,business.industry ,Colorectal cancer ,media_common.quotation_subject ,Mutant RAS ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Organoid ,Cancer research ,Medicine ,In patient ,business ,030215 immunology ,media_common - Published
- 2016
- Full Text
- View/download PDF
20. cAMP regulates DEP domain-mediated binding of the guanine nucleotide exchange factor Epac1 to phosphatidic acid at the plasma membrane
- Author
-
Sarah V. Consonni, Johannes L. Bos, Martijn Gloerich, and Emma Spanjaard
- Subjects
Conformational change ,Protein Conformation ,Phosphatidic Acids ,Small G Protein ,Biology ,chemistry.chemical_compound ,Cyclic AMP ,Guanine Nucleotide Exchange Factors ,Humans ,Multidisciplinary ,Cell Membrane ,rap1 GTP-Binding Proteins ,Phosphatidic acid ,Biological Sciences ,Lipids ,Recombinant Proteins ,Protein Structure, Tertiary ,Cell biology ,Pleckstrin homology domain ,Protein Transport ,HEK293 Cells ,chemistry ,Liposomes ,DEP domain ,CAMP binding ,sense organs ,Guanine nucleotide exchange factor ,Phospholipase D1 ,Protein Binding - Abstract
Epac1 is a cAMP-regulated guanine nucleotide exchange factor for the small G protein Rap. Upon cAMP binding, Epac1 undergoes a conformational change that results in its release from autoinhibition. In addition, cAMP induces the translocation of Epac1 from the cytosol to the plasma membrane. This relocalization of Epac1 is required for efficient activation of plasma membrane-located Rap and for cAMP-induced cell adhesion. This translocation requires the Dishevelled, Egl-10, Pleckstrin (DEP) domain, but the molecular entity that serves as the plasma membrane anchor and the possible mechanism of regulated binding remains elusive. Here we show that Epac1 binds directly to phosphatidic acid. Similar to the cAMP-induced Epac1 translocation, this binding is regulated by cAMP and requires the DEP domain. Furthermore, depletion of phosphatidic acid by inhibition of phospholipase D1 prevents cAMP-induced translocation of Epac1 as well as the subsequent activation of Rap at the plasma membrane. Finally, mutation of a single basic residue within a polybasic stretch of the DEP domain, which abolishes translocation, also prevents binding to phosphatidic acid. From these results we conclude that cAMP induces a conformational change in Epac1 that enables DEP domain-mediated binding to phosphatidic acid, resulting in the tethering of Epac1 at the plasma membrane and subsequent activation of Rap.
- Published
- 2012
- Full Text
- View/download PDF
21. Epac1 and PDZ-GEF cooperate in Rap1 mediated endothelial junction control
- Author
-
Emma Spanjaard, Jelena R. Linnemann, Johan de Rooij, Patricia M. Brouwer, Jantine J.G. van Dijk, On Ying A. Chan, Fried J. T. Zwartkruis, Holger Rehmann, Johannes L. Bos, Stephan Huveneers, Willem-Jan Pannekoek, Anne Jan van der Meer, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
rho GTP-Binding Proteins ,endocrine system ,Botulinum Toxins ,Telomere-Binding Proteins ,PDZ domain ,Motility ,Nerve Tissue Proteins ,Biology ,Time-Lapse Imaging ,Shelterin Complex ,Adherens junction ,Antigens, CD ,Live cell imaging ,Cyclic AMP ,Electric Impedance ,Human Umbilical Vein Endothelial Cells ,Guanine Nucleotide Exchange Factors ,Humans ,Barrier function ,Actin ,ADP Ribose Transferases ,HEK 293 cells ,Adherens Junctions ,Cell Biology ,Cadherins ,Actins ,Cell biology ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,Microscopy, Fluorescence ,Gene Knockdown Techniques ,RNA Interference ,Rap1 ,Endothelium, Vascular - Abstract
Epac1 and its effector Rap1 are important mediators of cAMP induced tightening of endothelial junctions and consequential increased barrier function. We have investigated the involvement of Rap1 signalling in basal, unstimulated, barrier function of a confluent monolayer of HUVEC using real time Electric Cell-substrate Impedance Sensing. Depletion of Rap1, but not Epac1, results in a strong decrease in barrier function. This decrease is also observed when cells are depleted of the cAMP independent Rap exchange factors PDZ-GEF1 and 2, showing that PDZ-GEFs are responsible for Rap1 activity in control of basal barrier function. Monolayers of cells depleted of PDZ-GEF or Rap1 show an irregular, zipper-like organization of VE-cadherin and live imaging of VE-cadherin-GFP reveals enhanced junction motility upon depletion of PDZ-GEF or Rap1. Importantly, activation of Epac1 increases the formation of cortical actin bundles at the cell-cell junctions, inhibits junction motility and restores barrier function of PDZ-GEFs depleted, but not Rap1 depleted cells. We conclude that PDZ-GEF activates Rap1 under resting conditions to stabilize cell-cell junctions and maintain basal integrity. Activation of Rap1 by cAMP/Epac1 induces junctional actin to further tighten cell-cell contacts.
- Published
- 2011
- Full Text
- View/download PDF
22. Evolution of the TOR Pathway
- Author
-
Johannes L. Bos, Fried J. T. Zwartkruis, Teunis J. P. van Dam, and Berend Snel
- Subjects
Rheb ,Protozoan Proteins ,Gene regulatory network ,Regulator ,P70-S6 Kinase 1 ,Biology ,Article ,Conserved sequence ,Evolution, Molecular ,Fungal Proteins ,Gene Duplication ,Genetics ,Animals ,Humans ,Gene Regulatory Networks ,Amino Acid Sequence ,AGC kinases ,Molecular Biology ,Transcription factor ,Conserved Sequence ,Phylogeny ,Ecology, Evolution, Behavior and Systematics ,TSC1-2 complex ,Fungal protein ,Pathway evolution ,Models, Genetic ,Sequence Homology, Amino Acid ,TOR Serine-Threonine Kinases ,TOR ,Gene Expression Regulation ,Multiprotein Complexes ,biology.protein ,Energy and redox metabolism Mitochondrial medicine [NCMLS 4] ,Signal Transduction ,Transcription Factors ,RHEB - Abstract
The TOR kinase is a major regulator of growth in eukaryotes. Many components of the TOR pathway are implicated in cancer and metabolic diseases in humans. Analysis of the evolution of TOR and its pathway may provide fundamental insight into the evolution of growth regulation in eukaryotes and provide a practical framework on which experimental evidence can be compared between species. Here we performed phylogenetic analyses on the components of the TOR pathway and determined their point of invention. We find that the two TOR complexes and a large part of the TOR pathway originated before the Last Eukaryotic Common Ancestor and form a core to which new inputs have been added during animal evolution. In addition, we provide insight into how duplications and sub-functionalization of the S6K, RSK, SGK and PKB kinases shaped the complexity of the TOR pathway. In yeast we identify novel AGC kinases that are orthologous to the S6 kinase. These results demonstrate how a vital signaling pathway can be both highly conserved and flexible in eukaryotes. Electronic supplementary material The online version of this article (doi:10.1007/s00239-011-9469-9) contains supplementary material, which is available to authorized users.
- Published
- 2011
23. Spatial Regulation of Cyclic AMP-Epac1 Signaling in Cell Adhesion by ERM Proteins
- Author
-
Matthijs R.H. Kooistra, Holger Rehmann, Fried J. T. Zwartkruis, Leo S. Price, Jun Zhao, Johannes L. Bos, Kees Jalink, Martijn Gloerich, Marjolein J. Vliem, Bas Ponsioen, Zhongchun Zhang, and Laila Ritsma
- Subjects
Small interfering RNA ,Recombinant Fusion Proteins ,Small G Protein ,Biology ,Cell Line ,Receptors, G-Protein-Coupled ,Cell membrane ,Radixin ,Two-Hybrid System Techniques ,Cell Adhesion ,Cyclic AMP ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Cell adhesion ,Molecular Biology ,Cell Membrane ,Microfilament Proteins ,Membrane Proteins ,rap1 GTP-Binding Proteins ,Articles ,Cell Biology ,Extracellular Matrix ,Cell biology ,Cytoskeletal Proteins ,medicine.anatomical_structure ,Membrane protein ,Guanine nucleotide exchange factor ,Signal transduction ,Signal Transduction - Abstract
Epac1 is a guanine nucleotide exchange factor for the small G protein Rap and is involved in membrane-localized processes such as integrin-mediated cell adhesion and cell-cell junction formation. Cyclic AMP (cAMP) directly activates Epac1 by release of autoinhibition and in addition induces its translocation to the plasma membrane. Here, we show an additional mechanism of Epac1 recruitment, mediated by activated ezrin-radixin-moesin (ERM) proteins. Epac1 directly binds with its N-terminal 49 amino acids to ERM proteins in their open conformation. Receptor-induced activation of ERM proteins results in increased binding of Epac1 and consequently the clustered localization of Epac1 at the plasma membrane. Deletion of the N terminus of Epac1, as well as disruption of the Epac1-ERM interaction by an interfering radixin mutant or small interfering RNA (siRNA)-mediated depletion of the ERM proteins, impairs Epac1-mediated cell adhesion. We conclude that ERM proteins are involved in the spatial regulation of Epac1 and cooperate with cAMP- and Rap-mediated signaling to regulate adhesion to the extracellular matrix.
- Published
- 2010
- Full Text
- View/download PDF
24. Epac: Defining a New Mechanism for cAMP Action
- Author
-
Johannes L. Bos and Martijn Gloerich
- Subjects
G protein ,Small G Protein ,Kidney ,Toxicology ,Capillary Permeability ,Glucagon-Like Peptide 1 ,Insulin Secretion ,Receptors, Adrenergic, beta ,Cyclic AMP ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Insulin ,Ion channel ,Inflammation ,Neurons ,Pharmacology ,Chemistry ,Biological activity ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,Mechanism of action ,Biochemistry ,Second messenger system ,Calcium ,Guanine nucleotide exchange factor ,medicine.symptom ,Signal transduction - Abstract
cAMP is a second messenger that is essential for relaying hormonal responses in many biological processes. The discovery of the cAMP target Epac explained various effects of cAMP that could not be attributed to the established targets PKA and cyclic nucleotide–gated ion channels. Epac1 and Epac2 function as guanine nucleotide exchange factors for the small G protein Rap. cAMP analogs that selectively activate Epac have helped to reveal a role for Epac in processes ranging from insulin secretion to cardiac contraction and vascular permeability. Advances in the understanding of the activation mechanism of Epac and its regulation by diverse anchoring mechanisms have helped to elucidate the means by which cAMP fulfills these functions via Epac.
- Published
- 2010
- Full Text
- View/download PDF
25. Changes in the signalling status of the small GTP-binding proteins Rac and Rho do not influence insulin-stimulated hexose transport
- Author
-
Johannes L. Bos, G. C. M. van der Zon, Johannes A Maassen, and J. Dorrestijn
- Subjects
G protein ,Endocrinology, Diabetes and Metabolism ,Biology ,Mice ,chemistry.chemical_compound ,Endocrinology ,GTP-binding protein regulators ,GTP-Binding Proteins ,Lysophosphatidic acid ,Adipocytes ,Internal Medicine ,Animals ,Hypoglycemic Agents ,Insulin ,Hexose ,Hexose transport ,Hexoses ,chemistry.chemical_classification ,Kinase ,Biological Transport ,3T3 Cells ,General Medicine ,Stimulation, Chemical ,rac GTP-Binding Proteins ,Cell biology ,Molecular Weight ,Insulin receptor ,chemistry ,Biochemistry ,biology.protein ,GLUT4 ,Signal Transduction - Abstract
Post-receptor signalling molecules that convey the signal from the activated insulin receptor to the actual process of Glut4 translocation and hexose uptake are poorly understood. Various studies have suggested a requirement of the lipid kinase phosphatidylinositol-3 kinase (P13-kinase) in this process. PI3kinase regulates the activation status of the small GTP-binding protein Rac which, in turn, is able to activate another G-protein Rho. Rac and Rho are known to regulate the structure of the membrane- and cytoplasmic actin-cytoskeleton. We have examined whether Rac and Rho transfer the signals generated by PI3kinase towards insulin-stimulated hexose uptake. For that purpose, we expressed in 3T3-L1 adipocytes the dominant-negative mutant of Rac N17 using vaccinia virus-mediated gene transfer. The expression levels of the Rac N17 protein were monitored by Western blotting. The abrogation of endogenous Rac signalling by expression of Rac N17 was inferred from the observed loss of arachidonic acid release in response to insulin. Basal and insulin-stimulated hexose transport were not affected by expression of the Rac Nl7 mutant. A possible contribution of Rho.GTP to stimulation of hexose uptake was examined by pre-incubation of adipocytes with lysophosphatidic acid (LPA). We observed a profound effect of LPA on the structure of the cytoskeleton and on the phosphorylation of Focal Adhesion Kinase (p125 FAK ), indicating that 3T3-L1 adipocytes respond to LPA and that Rho was activated by LPA. However, no effect was detected on the basal or on the insulin-stimulated hexose transport. We conclude that Rac and Rho are unlikely to be involved in insulin-stimulated hexose transport, suggesting a possible contribution of other signalling pathways, downstream of PI3kinase to this process.
- Published
- 2009
- Full Text
- View/download PDF
26. Direct Spatial Control of Epac1 by Cyclic AMP
- Author
-
Johannes L. Bos, Martijn Gloerich, Holger Rehmann, Laila Ritsma, Kees Jalink, and Bas Ponsioen
- Subjects
Conformational change ,Recombinant Fusion Proteins ,Rap GTP-binding protein ,Biology ,Cell Line ,Cell membrane ,chemistry.chemical_compound ,Cell Adhesion ,Cyclic AMP ,medicine ,Guanine Nucleotide Exchange Factors ,Humans ,Cell adhesion ,Molecular Biology ,Total internal reflection fluorescence microscope ,Forskolin ,Cell Membrane ,Colforsin ,fungi ,Isoproterenol ,Articles ,Cell Biology ,Adrenergic beta-Agonists ,Cell biology ,rap GTP-Binding Proteins ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,DEP domain ,CAMP binding ,sense organs ,Signal Transduction - Abstract
Epac1 is a guanine nucleotide exchange factor (GEF) for the small G protein Rap and is directly activated by cyclic AMP (cAMP). Upon cAMP binding, Epac1 undergoes a conformational change that allows the interaction of its GEF domain with Rap, resulting in Rap activation and subsequent downstream effects, including integrin-mediated cell adhesion and cell-cell junction formation. Here, we report that cAMP also induces the translocation of Epac1 toward the plasma membrane. Combining high-resolution confocal fluorescence microscopy with total internal reflection fluorescence and fluorescent resonance energy transfer assays, we observed that Epac1 translocation is a rapid and reversible process. This dynamic redistribution of Epac1 requires both the cAMP-induced conformational change as well as the DEP domain. In line with its translocation, Epac1 activation induces Rap activation predominantly at the plasma membrane. We further show that the translocation of Epac1 enhances its ability to induce Rap-mediated cell adhesion. Thus, the regulation of Epac1-Rap signaling by cAMP includes both the release of Epac1 from autoinhibition and its recruitment to the plasma membrane.
- Published
- 2009
- Full Text
- View/download PDF
27. The RapGEF PDZ-GEF2 is required for maturation of cell–cell junctions
- Author
-
Judith Klumperman, Nadia Dubé, Johannes L. Bos, Marjolein J. Vliem, Willem-Jan Pannekoek, Viola Oorschot, Holger Rehmann, and Matthijs R.H. Kooistra
- Subjects
Chemistry ,Activator (genetics) ,Cadherin ,PDZ domain ,Regulator ,rap1 GTP-Binding Proteins ,Epithelial Cells ,Adherens Junctions ,Cell Biology ,Cadherins ,Cell junction ,Cell biology ,Adherens junction ,rap GTP-Binding Proteins ,Cell Adhesion ,Guanine Nucleotide Exchange Factors ,Humans ,Rap1 ,Endothelium, Vascular ,Guanine nucleotide exchange factor - Abstract
The small G-protein Rap1 is a critical regulator of cell–cell contacts and is activated by the remodeling of adherens junctions. Here we identify the Rap1 guanine nucleotide exchange factor PDZ-GEF2 as an upstream activator of Rap1 required for the maturation of adherens junctions in the lung carcinoma cells A549. Knockdown of PDZ-GEF2 results in the persistence of adhesion zippers at cell–cell contacts. Activation of Rap1A rescues junction maturation in absence of PDZ-GEF2, demonstrating that Rap1A is downstream of PDZ-GEF2 in this process. Moreover, depletion of Rap1A, but not Rap1B, impairs adherens junction maturation. siRNA for PDZ-GEF2 also lowers the levels of E-cadherin, an effect that can be mimicked by Rap1B, but not Rap1A siRNA. Since junctions in Rap1B depleted cells have a mature appearance, these data suggest that PDZ-GEF2 activates Rap1A and Rap1B to perform different functions. Our results present the first direct evidence that PDZ-GEF2 plays a critical role in the maturation of adherens junctions.
- Published
- 2008
- Full Text
- View/download PDF
28. PKA and Epac1 regulate endothelial integrity and migration through parallel and independent pathways
- Author
-
Peter L. Hordijk, Magdalena J. Lorenowicz, Johannes L. Bos, Matthijs R.H. Kooistra, Mar Fernandez-Borja, Landsteiner Laboratory, and Physiology
- Subjects
Integrins ,Histology ,Endothelium ,Telomere-Binding Proteins ,Integrin ,Motility ,Transfection ,Shelterin Complex ,Pathology and Forensic Medicine ,Capillary Permeability ,Cell Movement ,Cyclic AMP ,Electric Impedance ,medicine ,Guanine Nucleotide Exchange Factors ,Humans ,Cells, Cultured ,Focal Adhesions ,biology ,Cell migration ,Cell Biology ,General Medicine ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,Enzyme Activation ,Endothelial stem cell ,Vascular endothelial growth factor A ,medicine.anatomical_structure ,biology.protein ,Rap1 ,Endothelium, Vascular ,Signal transduction ,Signal Transduction - Abstract
The vascular endothelium provides a semi-permeable barrier, which restricts the passage Of fluid, macromolecules and cells to the surrounding tissues. Cyclic AMP promotes endothelial barrier function and protects the endothelium against pro-inflammatory mediators. This study analyzed the relative contribution of two cAMP targets, PKA and Epac1, to the control of endothelial barrier function and endothelial cell migration. Real-time recording of transendothelial electrical resistance showed that activation of either PKA or Epac1 with specific cAMP analogues increases endothelial barrier function and promotes endothelial cell migration. In addition, reduction of Epac1 expression showed that Epac1 and PKA control endothelial integrity and cell motility by two independent and complementary signaling pathways. We demonstrate that integrin-mediated adhesion is required for PKA, but not Epac1-Rap1-driven stimulation or endothelial barrier function. In contrast, both PKA- and Epac1-stimulated endothelial cell migration requires integrin function. These data show that activation of Epac1 and PKA by cAMP results in the stimulation of two parallel, independent signaling pathways that positively regulate endothelial integrity and cell migration, which is important for recovery after endothelial damage and for restoration of compromised endothelial barrier function. (C) 2008 Elsevier GmbH. All rights reserved
- Published
- 2008
- Full Text
- View/download PDF
29. Capturing cyclic nucleotides in action: snapshots from crystallographic studies
- Author
-
Holger Rehmann, Johannes L. Bos, and Alfred Wittinghofer
- Subjects
chemistry.chemical_classification ,Chemistry ,Guanine ,Receptors, Cell Surface ,Cell Biology ,Crystallography, X-Ray ,Ion Channels ,Cell biology ,chemistry.chemical_compound ,Biochemistry ,Cyclic nucleotide binding ,Second messenger system ,Cyclic AMP ,Animals ,Humans ,Nucleotide ,Signal transduction ,Protein kinase A ,Cyclic GMP ,Protein Kinases ,Molecular Biology ,cGMP-dependent protein kinase ,Ion channel - Abstract
Fifty years ago, cyclic AMP was discovered as a second messenger of hormone action, heralding the age of signal transduction. Many cellular processes were found to be regulated by cAMP and the related cyclic GMP. Cyclic nucleotides function by binding to and activating their effectors - protein kinase A, protein kinase G, cyclic-nucleotide-regulated ion channels and the guanine nucleotide-exchange factor Epac. Recent structural insights have now made it possible to propose a general structural mechanism for how cyclic nucleotides regulate these proteins.
- Published
- 2007
- Full Text
- View/download PDF
30. cAMP sensor Epac as a determinant of ATP-sensitive potassium channel activity in human pancreatic β cells and rat INS-1 cells
- Author
-
Michael J. Rindler, Guoxin Kang, Brian Malester, Frank Schwede, Holger Rehmann, George G. Holz, William A. Coetzee, Oleg G. Chepurny, and Johannes L. Bos
- Subjects
Membrane potential ,ATP-sensitive potassium channel ,Biochemistry ,Physiology ,HEK 293 cells ,Second messenger system ,Sulfonylurea receptor ,Depolarization ,Biology ,Protein kinase A ,Potassium channel ,Cell biology - Abstract
The Epac family of cAMP-regulated guanine nucleotide exchange factors (cAMPGEFs, also known as Epac1 and Epac2) mediate stimulatory actions of the second messenger cAMP on insulin secretion from pancreatic beta cells. Because Epac2 is reported to interact in vitro with the isolated nucleotide-binding fold-1 (NBF-1) of the beta-cell sulphonylurea receptor-1 (SUR1), we hypothesized that cAMP might act via Epac1 and/or Epac2 to inhibit beta-cell ATP-sensitive K+ channels (K(ATP) channels; a hetero-octomer of SUR1 and Kir6.2). If so, Epac-mediated inhibition of K(ATP) channels might explain prior reports that cAMP-elevating agents promote beta-cell depolarization, Ca2+ influx and insulin secretion. Here we report that Epac-selective cAMP analogues (2'-O-Me-cAMP; 8-pCPT-2'-O-Me-cAMP; 8-pMeOPT-2'-O-Me-cAMP), but not a cGMP analogue (2'-O-Me-cGMP), inhibit the function of K(ATP) channels in human beta cells and rat INS-1 insulin-secreting cells. Inhibition of K(ATP) channels is also observed when cAMP, itself, is administered intracellularly, whereas no such effect is observed upon administration N6-Bnz-cAMP, a cAMP analogue that activates protein kinase A (PKA) but not Epac. The inhibitory actions of Epac-selective cAMP analogues at K(ATP) channels are mimicked by a cAMP agonist (8-Bromoadenosine-3', 5'-cyclic monophosphorothioate, Sp-isomer, Sp-8-Br-cAMPS), but not a cAMP antagonist (8-Bromoadenosine-3', 5'-cyclic monophosphorothioate, Rp-isomer, Rp-8-Br-cAMPS), and are abrogated following transfection of INS-1 cells with a dominant-negative Epac1 that fails to bind cAMP. Because both Epac1 and Epac2 coimmunoprecipitate with full-length SUR1 in HEK cell lysates, such findings delineate a novel mechanism of second messenger signal transduction in which cAMP acts via Epac to modulate ion channel function, an effect measurable as the inhibition of K(ATP) channel activity in pancreatic beta cells.
- Published
- 2006
- Full Text
- View/download PDF
31. Amino acids mediate mTOR/raptor signaling through activation of class 3 phosphatidylinositol 3OH-kinase
- Author
-
Manel Joaquin, So Young Kim, Marta Roccio, Jonathan M. Backer, Fried J. T. Zwartkruis, Francois Natt, Johannes L. Bos, Takahiro Nobukuni, George Thomas, Stephen G. Dann, Pawan Gulati, and Maya P. Byfield
- Subjects
medicine.medical_treatment ,Blotting, Western ,Biology ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Cell Line, Tumor ,medicine ,Humans ,Phosphatidylinositol ,Amino Acids ,RNA, Small Interfering ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Monomeric GTP-Binding Proteins ,chemistry.chemical_classification ,Multidisciplinary ,Kinase ,TOR Serine-Threonine Kinases ,Insulin ,Neuropeptides ,RPTOR ,Proteins ,Regulatory-Associated Protein of mTOR ,Biological Sciences ,Amino acid ,Cell biology ,Microscopy, Fluorescence ,chemistry ,Biochemistry ,Ras Homolog Enriched in Brain Protein ,Signal transduction ,Protein Kinases ,Signal Transduction ,Hormone - Abstract
During the evolution of metazoans and the rise of systemic hormonal regulation, the insulin-controlled class 1 phosphatidylinositol 3OH-kinase (PI3K) pathway was merged with the primordial amino acid-driven mammalian target of rapamycin (mTOR) pathway to control the growth and development of the organism. Insulin regulates mTOR function through a recently described canonical signaling pathway, which is initiated by the activation of class 1 PI3K. However, how the amino acid input is integrated with that of the insulin signaling pathway is unclear. Here we used a number of molecular, biochemical, and pharmacological approaches to address this issue. Unexpectedly, we found that a major pathway by which amino acids control mTOR signaling is distinct from that of insulin and that, instead of signaling through components of the insulin/class 1 PI3K pathway, amino acids mediate mTOR activation by signaling through class 3 PI3K, hVps34.
- Published
- 2005
- Full Text
- View/download PDF
32. Epac1 regulates integrity of endothelial cell junctions through VE-cadherin
- Author
-
Johannes L. Bos, Matthijs R.H. Kooistra, Monica Corada, and Elisabetta Dejana
- Subjects
Biophysics ,Biology ,Biochemistry ,Cell junction ,Adherens junction ,chemistry.chemical_compound ,Antigens, CD ,Structural Biology ,cAMP ,Cyclic AMP ,Genetics ,Guanine Nucleotide Exchange Factors ,Humans ,RNA, Small Interfering ,Cytoskeleton ,GTPase ,Molecular Biology ,Cells, Cultured ,Actin ,Forskolin ,Endothelial Cells ,rap1 GTP-Binding Proteins ,Actin remodeling ,Cell Biology ,Cadherins ,Actin cytoskeleton ,Actins ,Cell biology ,Enzyme Activation ,Endothelial stem cell ,Intercellular Junctions ,chemistry ,RNAi ,Endothelium, Vascular ,permeability - Abstract
We have previously shown that Rap1 as well as its guanine nucleotide exchange factor Epac1 increases cell–cell junction formation. Here, we show that activation of Epac1 with the exchange protein directly activated by cAMP (Epac)-specific cAMP analog 8CPT-2′O-Me-cAMP (007) resulted in a tightening of the junctions and a decrease in the permeability of the endothelial cell monolayer. In addition, 007 treatment resulted in the breakdown of actin stress fibers and the formation of cortical actin. These effects were completely inhibited by siRNA against Epac1. In VE-cadherin knock-out cells Epac1 did not affect cell permeability, whereas in cells re-expressing VE-cadherin this effect was restored. Finally, the effect of Epac activation on the actin cytoskeleton was independent of junction formation. From these results we conclude that in human umbilical vein endothelial cells Epac1 controls VE-cadherin-mediated cell junction formation and induces reorganization of the actin cytoskeleton.
- Published
- 2005
- Full Text
- View/download PDF
33. Down-regulation of Rap1 activity is involved in ephrinB1-induced cell contraction
- Author
-
Hans Clevers, Dominique T. Brandt, Eduard Batlle, Leo S. Price, Johannes L. Bos, and Jurgen Riedl
- Subjects
rho GTP-Binding Proteins ,Colon ,Receptor, EphB2 ,Cell ,Down-Regulation ,Ephrin-B1 ,Biology ,Biochemistry ,Cell Movement ,Cell Line, Tumor ,Precursor cell ,Cell Adhesion ,medicine ,Humans ,Ephrin ,Cell adhesion ,Cell Shape ,Molecular Biology ,digestive, oral, and skin physiology ,Erythropoietin-producing hepatocellular (Eph) receptor ,rap1 GTP-Binding Proteins ,Cell Biology ,Molecular biology ,Cell biology ,medicine.anatomical_structure ,Gene Expression Regulation ,ras GTPase-Activating Proteins ,Cell culture ,Rap1 ,Tyrosine kinase ,Research Article - Abstract
Ephrins are cell surface ligands that activate Eph receptor tyrosine kinases. This ligand–receptor interaction plays a central role in the sorting of cells. We have previously shown that the ephrinB–EphB signalling pathway is also involved in the migration of intestinal precursor cells along the crypts. Using the colon cell line DLD1 expressing the EphB2 receptor, we showed that stimulation of these cells with soluble ephrinB1 results in a rapid retraction of cell extensions and a detachment of cells. On ephrinB1 stimulation, the small GTPases Rho and Ras are activated and Rap1 is inactivated. Importantly, when a constitutively active Rap1 mutant was introduced into these cells, ephrinB1-induced retraction was inhibited. From these results, we conclude that down-regulation of Rap1 is a prerequisite for ephrin-induced cell retraction in colon cells.
- Published
- 2005
- Full Text
- View/download PDF
34. The cAMP-Epac-Rap1 Pathway Regulates Cell Spreading and Cell Adhesion to Laminin-5 through the α3β1 Integrin but Not the α6β4 Integrin
- Author
-
Jorrit M. Enserink, Trond Methi, Leo S. Price, Johannes L. Bos, Milada Mahic, Arnoud Sonnenberg, and Kjetil Taskén
- Subjects
DNA, Complementary ,Time Factors ,Blotting, Western ,Green Fluorescent Proteins ,Integrin ,Cell Separation ,Transfection ,Biochemistry ,Focal adhesion ,Cell Movement ,Laminin ,Cell Line, Tumor ,Cell Adhesion ,Cyclic AMP ,Guanine Nucleotide Exchange Factors ,Humans ,Phosphorylation ,Cell adhesion ,Molecular Biology ,Integrin alpha6beta4 ,biology ,Chemistry ,Cell adhesion molecule ,Integrin alpha3beta1 ,rap1 GTP-Binding Proteins ,Cell Biology ,Flow Cytometry ,Intercellular adhesion molecule ,Actins ,Extracellular Matrix ,Cell biology ,Fibronectin ,biology.protein ,Neural cell adhesion molecule ,K562 Cells ,Cell Adhesion Molecules ,Protein Binding ,Signal Transduction - Abstract
Laminin-5 is an important constituent of the basal lamina. The receptors for laminin-5, the integrins alpha3beta1 and alpha6beta4, have been associated with epithelial wound migration and carcinoma invasion. The signal transduction mechanisms that regulate these integrins are not well understood. We report here that the small GTPase Rap1 regulates the adhesion of a number of cell lines to various extracellular matrix proteins including laminin-5. cAMP also mediates cell adhesion and spreading on laminin-5, a process that is independent of protein kinase A but rather dependent on Epac1, a cAMP-dependent exchange factor for Rap. Interestingly, although both alpha3beta1 and alpha6beta4 mediate adhesion to laminin-5, only alpha3beta1-dependent adhesion is dependent on Rap1. These results provide evidence for a function of the cAMP-Epac-Rap1 pathway in cell adhesion and spreading on different extracellular matrix proteins. They also define different roles for the laminin-binding integrins in regulated cell adhesion and subsequent cell spreading.
- Published
- 2004
- Full Text
- View/download PDF
35. Molecular mechanisms in signal transduction and cancer
- Author
-
Boudewijn M.T. Burgering and Johannes L. Bos
- Subjects
Genetics ,Senescence ,Cancer ,Growth control ,Reproductive age ,Biology ,medicine.disease ,Biochemistry ,Cancer cell ,medicine ,Stem cell ,Signal transduction ,Older people ,Molecular Biology - Abstract
The EMBL/Salk/EMBO Conference on ‘Oncogenes & Growth Control’ was held at the EMBL in Heidelberg, Germany, between 17 and 20 April 2004, and was organized by M. Bienz, G. Evan, C. Marshall, A. Nebreda and R. Treisman ![][1] Good weather and a pint of apple juice (although we thought we had ordered beer) at a farmhouse opposite the European Molecular Biology Laboratory (EMBL) heralded the start of the 15th conference on ‘Oncogenes & Growth Control’. The participants had the oppor‐tunity to enjoy the friendly atmosphere at the EMBL, the hospitality of the staff and the tasty food. And of course, a series of excellent talks, which provided an in‐depth view of several topics in signal trans‐duction and cancer biology. In this report, we will give you an overview of what was discussed. ### Starting at the end In the last session of the meeting, J. Campisi (Berkeley, CA, USA) looked at cancer from an evolutionary point of view and presented a model in which cancer is not only caused by the accumulation of mutations, but also facilitated by the senescent cells that accumulate in older people. Her model is based on the observation that senescent cells support the growth of preneoplastic cells much better than that of normal fibroblasts. Senescent cells may do so by secreting many products, including growth factors, cytokines such as interleukin‐6 (IL‐6), and proteases. Older people do indeed accumulate senescent cells as shown by β‐galactosidase staining. Furthermore, older people have higher levels of IL‐6, which is one of the causes of frailty. Campisi argued that the occurrence of senescent cells is a pleiotropic effect of our fight against cancer cells, and as they occur after reproductive age, there is no selective pressure to eliminate them. This fight against cancer has also resulted in the evolution of tumour suppressor genes, which fall into … [1]: /embed/graphic-1.gif
- Published
- 2004
- Full Text
- View/download PDF
36. Rap1 Signaling Is Required for Suppression of Ras-Generated Reactive Oxygen Species and Protection Against Oxidative Stress in T Lymphocytes
- Author
-
Fried J. T. Zwartkruis, Philip H. J. Remans, Ferdinand C. Breedveld, Paul P. Tak, Ellen A. M. Papendrecht-van der Voort, Sonja I. Gringhuis, Johannes L. Bos, Kris A. Reedquist, Jacob M van Laar, E. W. Nivine Levarht, Marcela Rosas, M E Sanders, Cornelis L. Verweij, Paul J. Coffer, Clinical Immunology and Rheumatology, Other departments, Amsterdam institute for Infection and Immunity, and Experimental Immunology
- Subjects
Time Factors ,T-Lymphocytes ,T cell ,Immunology ,Biology ,medicine.disease_cause ,Jurkat cells ,Jurkat Cells ,Phosphatidylinositol 3-Kinases ,Anti-apoptotic Ras signalling cascade ,medicine ,Humans ,Immunology and Allergy ,Small GTPase ,T-cell receptor ,rap1 GTP-Binding Proteins ,Cell biology ,Oxidative Stress ,medicine.anatomical_structure ,ras Proteins ,Rap1 ,Mitogen-Activated Protein Kinases ,Signal transduction ,Reactive Oxygen Species ,Oxidative stress ,Signal Transduction - Abstract
Transient production of reactive oxygen species (ROS) plays an important role in optimizing transcriptional and proliferative responses to TCR signaling in T lymphocytes. Conversely, chronic oxidative stress leads to decreased proliferative responses and enhanced transcription of inflammatory gene products, and is thought to underlie the altered pathogenic behavior of T lymphocytes in some human diseases, such as rheumatoid arthritis (RA). Although the signaling mechanisms regulating ROS production in T lymphocytes has not been identified, activation of the small GTPase Ras has been shown to couple agonist stimulation to ROS production in other cell types. We find that Ras signaling via Ral stimulates ROS production in human T lymphocytes, and is required for TCR and phorbol ester-induced ROS production. The related small GTPase Rap1 suppresses agonist, Ras and Ral–dependent ROS production through a PI3K–dependent pathway, identifying a novel mechanism by which Rap1 can distally antagonize Ras signaling pathways. In synovial fluid T lymphocytes from RA patients we observed a high rate of endogenous ROS production, correlating with constitutive Ras activation and inhibition of Rap1 activation. Introduction of dominant-negative Ras into synovial fluid T cells restored redox balance, providing evidence that deregulated Ras and Rap1 signaling underlies oxidative stress and consequent altered T cell function observed in RA.
- Published
- 2004
- Full Text
- View/download PDF
37. Regulation of sterol carrier protein gene expression by the Forkhead transcription factor FOXO3a
- Author
-
Johannes L. Bos, Nannette Jelluma, Karel W. A. Wirtz, Tobias B. Dansen, Ronald J.A. Wanders, Geert J. P. L. Kops, Simone Denis, Boudewijn M.T. Burgering, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases, and Paediatric Metabolic Diseases
- Subjects
QD415-436 ,Biology ,Biochemistry ,Cell Line ,Endocrinology ,Transcriptional regulation ,Humans ,oxidative stress ,peroxisome ,RNA, Messenger ,Acetyl-CoA C-Acetyltransferase ,Transcription factor ,fatty acid oxidation ,SCP2 ,Thiolase ,Forkhead Box Protein O1 ,Cell Cycle ,Forkhead Box Protein O3 ,aging ,Promoter ,Forkhead Transcription Factors ,Cell Biology ,Peroxisome ,DNA-Binding Proteins ,Sterol carrier protein ,Gene Expression Regulation ,Fatty acid analog ,Lipid Peroxidation ,Carrier Proteins ,Oxidation-Reduction ,Transcription Factors - Abstract
The SCP gene encodes two proteins, sterol carrier protein X (SCPx) and SCP2, that are independently regulated by separate promoters. SCPx has been shown to be the thiolase involved in the breakdown of branched-chain fatty acids and in the biosynthesis of bile acids. The in vivo function of SCP2 however remains to be established. The transcriptional regulation of SCPx and SCP2 is unclear, but their promoter regions contain several putative regulatory domains. We show here that both SCPx and SCP2 are upregulated by the daf-16-like Forkhead transcription factor FOXO3a (also known as FKHRL1) on the level of promoter activity. It was recently described that Forkheads regulate protection against (oxidative) stress in both Caenorhabditis elegans and mammalian cells. We looked into a role for SCP2 in the cellular defense against oxidative damage and found that a fluorescent fatty acid analog bound to SCP2 is protected against H2O2/Cu2+-induced oxidative damage. We propose a model for the way in which SCP2 could protect fatty acids from peroxidation.
- Published
- 2004
38. Ras Association-Domain Dimers Bring Proteins Together
- Author
-
Johannes L. Bos and Holger Rehmann
- Subjects
0301 basic medicine ,endocrine system ,Chemistry ,Association (object-oriented programming) ,Dimer ,Complex formation ,Small G Protein ,Cell biology ,enzymes and coenzymes (carbohydrates) ,03 medical and health sciences ,Crystallography ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Structural Biology ,Monomeric GTP-Binding Proteins ,Domain (ring theory) ,Journal Article ,Rap1 ,Signal transduction ,Molecular Biology ,030217 neurology & neurosurgery - Abstract
In this issue of Structure, Gingras et al. (2016) show that Ras association (RA) domains of the Rap1 and Ras interacting protein Rasip1 can form a dimer in the presence and absence of the small G protein Rap1. This provides an explanation for the observed complex formation in Rap1-mediated signaling.
- Published
- 2016
- Full Text
- View/download PDF
39. Ligand-mediated Activation of the cAMP-responsive Guanine Nucleotide Exchange Factor Epac
- Author
-
Alfred Wittinghofer, Holger Rehmann, Johannes L. Bos, Stein Ove Døskeland, and Frank Schwede
- Subjects
Recombinant Fusion Proteins ,Amino Acid Motifs ,DNA Mutational Analysis ,Plasma protein binding ,Calorimetry ,Ligands ,medicine.disease_cause ,Biochemistry ,Catalysis ,Cyclic nucleotide ,chemistry.chemical_compound ,Protein structure ,Cyclic AMP ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Nucleotide ,Protein kinase A ,Molecular Biology ,Glutathione Transferase ,chemistry.chemical_classification ,Mutation ,Dose-Response Relationship, Drug ,Chemistry ,Mutagenesis ,rap1 GTP-Binding Proteins ,Cell Biology ,Cyclic AMP-Dependent Protein Kinases ,Protein Structure, Tertiary ,Cell biology ,Kinetics ,Models, Chemical ,Spectrophotometry ,Guanine nucleotide exchange factor ,Protein Binding - Abstract
Epac is a cAMP-dependent exchange factor for the small GTP-binding protein Rap. The activity of Epac is inhibited by a direct interaction between the C-terminal helical part of the cAMP-binding domain, called the lid, and the catalytic region, which is released after binding of cAMP. Herein, we show that the activation properties are very sensitive to modifications of the cyclic nucleotide. Some analogues are inhibitory and others are stimulatory; some are characterized by a much higher activation potential than normal cAMP. Mutational analysis of Epac allows insights into a network of interactions between the cyclic nucleotides and Epac. Mutations in the lid region are able to amplify or to attenuate selectively the activation potency of cAMP analogues. The properties of cAMP analogues previously used for the activation of the cAMP responsive protein kinase A and of 8-(4-chlorophenylthio)-2'-O-methyladenosine-3',5'-cyclicmonophosphate, an analogue highly selective for activation of Epac were investigated in detail.
- Published
- 2003
- Full Text
- View/download PDF
40. cAMP Analog Mapping of Epac1 and cAMP Kinase
- Author
-
Carine Maenhaut, Johannes L. Bos, Frank Schwede, Stein Ove Døskeland, Hans G. Genieser, Aurora Martinez, Sarah Dremier, Johan de Rooij, Anne Elisabeth Christensen, Frode Selheim, and Khanh K. Dao
- Subjects
Neurite ,Chemistry ,Kinase ,Cell ,Cell Biology ,Biochemistry ,Cell biology ,chemistry.chemical_compound ,medicine.anatomical_structure ,Ribose ,medicine ,Guanine nucleotide exchange factor ,Binding site ,Protein kinase A ,Molecular Biology ,Binding selectivity - Abstract
Little is known about the relative role of cAMP-dependent protein kinase (cAPK) and guanine exchange factor directly activated by cAMP (Epac) as mediators of cAMP action. We tested cAMP analogs for ability to selectively activate Epac1 or cAPK and discriminate between the binding sites of Epac and of cAPKI and cAPKII. We found that commonly used cAMP analogs, like 8-Br-cAMP and 8-pCPT-cAMP, activate Epac and cAPK equally as well as cAMP, i.e. were full agonists. In contrast, 6-modified cAMP analogs, like N6-benzoyl-cAMP, were inefficient Epac activators and full cAPK activators. Analogs modified in the 2′-position of the ribose induced stronger Epac1 activation than cAMP but were only partial agonists for cAPK. 2′-O-Alkyl substitution of cAMP improved Epac/cAPK binding selectivity 10–100-fold. Phenylthio substituents in position 8, particularly with MeO- or Cl- in p-position, enhanced the Epac/cAPK selectivity even more. The combination of 8-pCPT- and 2′-O-methyl substitutions improved the Epac/cAPK binding selectivity about three orders of magnitude. The cAPK selectivity of 6-substituted cAMP analogs, the preferential inhibition of cAPK by moderate concentrations of Rp-cAMPS analogs, and the Epac selectivity of 8-pCPT-2′-O-methyl-cAMP was also demonstrated in intact cells. Using these compounds to selectively modulate Epac and cAPK in PC-12 cells, we observed that analogs selectively activating Epac synergized strongly with cAPK specific analogs to induce neurite outgrowth. We therefore conclude that cAMP-induced neurite outgrowth is mediated by both Epac and cAPK.
- Published
- 2003
- Full Text
- View/download PDF
41. Epac: a new cAMP target and new avenues in cAMP research
- Author
-
Johannes L. Bos
- Subjects
Camp analogue ,Chemistry ,Mechanism (biology) ,Cell Biology ,GTPase ,Bioinformatics ,Protein Structure, Tertiary ,Cell biology ,Protein structure ,Insulin Secretion ,Cell Adhesion ,Cyclic AMP ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Insulin ,Rap1 ,Guanine nucleotide exchange factor ,Insulin secretion ,Molecular Biology ,Insulin metabolism - Abstract
Five years ago, Epac--a guanine nucleotide exchange factor for the Ras-like small GTPases Rap1 and Rap2--was found to be a new target of cyclic AMP, which opened up new avenues for cAMP research. Structural analysis of the cAMP-binding domains of Epac2 has identified a unifying mechanism for how cAMP activates proteins, and the design and synthesis of an Epac-specific cAMP analogue has paved the way for future discoveries.
- Published
- 2003
- Full Text
- View/download PDF
42. The Small GTPase Rap1 Is Activated by Turbulence and Is Involved in Integrin αIIbβ3-mediated Cell Adhesion in Human Megakaryocytes
- Author
-
Jan-Willem N. Akkerman, Fried J. T. Zwartkruis, Kim M.T. de Bruyn, Johan de Rooij, and Johannes L. Bos
- Subjects
Blood Platelets ,endocrine system ,Integrin ,Platelet Glycoprotein GPIIb-IIIa Complex ,Transfection ,Biochemistry ,Jurkat cells ,Cell Line ,Jurkat Cells ,Cell surface receptor ,Cell Adhesion ,Tumor Cells, Cultured ,Humans ,Enzyme Inhibitors ,Cell adhesion ,Egtazic Acid ,Molecular Biology ,Protein Kinase C ,Protein kinase C ,Chelating Agents ,biology ,Cell adhesion molecule ,Fibrinogen ,rap1 GTP-Binding Proteins ,Cell Biology ,Actin cytoskeleton ,Recombinant Proteins ,Cell biology ,enzymes and coenzymes (carbohydrates) ,biology.protein ,Rap1 ,Stress, Mechanical ,K562 Cells ,Megakaryocytes ,Signal Transduction - Abstract
The small GTPase Rap1, which is activated by a large variety of stimuli, functions in the control of integrin-mediated cell adhesion. Here we show that in human megakaryocytes and several other commonly used hematopoietic cell lines such as K562, Jurkat, and THP-1, stress induced by gentle tumbling of the samples resulted in rapid and strong activation of Rap1. This turbulence-induced activation could not be blocked by inhibitors previously shown to affect Rap1 activation in human platelets, such as the intracellular calcium chelator BAPTA-AM (1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid) and various protein kinase C inhibitors. Also inhibition of actin cytoskeleton dynamics did not influence this activation of Rap1, suggesting that this activation is mediated by cell surface receptors. Human platelets, however, were refractory to turbulence-induced activation of Rap1. To determine the consequences of Rap1 activation we measured adhesion of megakaryocytes to fibrinogen, which is mediated by the integrin alphaIIbbeta3, in the presence of inhibitors of Rap1 signaling. Introduction of both Rap1GAP and RalGDS-RBD in the megakaryoblastic cell line DAMI strongly reduced basal adhesion to immobilized fibrinogen. This inhibition was partially rescued by the phorbol ester 12-O-tetradecanoylphorbol-13-acetate but not by alpha-thrombin. From these results we conclude that in megakaryocytes turbulence induces Rap1 activation that controls alphaIIbbeta3-mediated cell adhesion.
- Published
- 2003
- Full Text
- View/download PDF
43. Epac-selective cAMP Analog 8-pCPT-2′-O-Me-cAMP as a Stimulus for Ca2+-induced Ca2+ Release and Exocytosis in Pancreatic β-Cells
- Author
-
Michael B. Wheeler, Marie E. Monaco, Johannes L. Bos, George G. Holz, Hans-G. Genieser, Frank Schwede, Oleg G. Chepurny, Jamie W. Joseph, and Guoxin Kang
- Subjects
medicine.medical_specialty ,G protein ,Biology ,Biochemistry ,Exocytosis ,Article ,Islets of Langerhans ,Bacterial Proteins ,Internal medicine ,Cyclic AMP ,Electrochemistry ,medicine ,Guanine Nucleotide Exchange Factors ,Humans ,Protein kinase A ,Molecular Biology ,Cells, Cultured ,Effector ,Cell Biology ,Immunohistochemistry ,Cell biology ,Luminescent Proteins ,Endocrinology ,Second messenger system ,Calcium ,Rap1 ,Guanine nucleotide exchange factor ,Signal transduction - Abstract
The second messenger cAMP exerts powerful stimulatory effects on Ca(2+) signaling and insulin secretion in pancreatic beta-cells. Previous studies of beta-cells focused on protein kinase A (PKA) as a downstream effector of cAMP action. However, it is now apparent that cAMP also exerts its effects by binding to cAMP-regulated guanine nucleotide exchange factors (Epac). Although one effector of Epac is the Ras-related G protein Rap1, it is not fully understood what the functional consequences of Epac-mediated signal transduction are at the cellular level. 8-(4-chloro-phenylthio)-2'-O-methyladenosine-3'-5'-cyclic monophosphate (8-pCPT-2'-O-Me-cAMP) is a newly described cAMP analog, and it activates Epac but not PKA. Here we demonstrate that 8-pCPT-2'-O-Me-cAMP acts in human pancreatic beta-cells and INS-1 insulin-secreting cells to mobilize Ca(2+) from intracellular Ca(2+) stores via Epac-mediated Ca(2+)-induced Ca(2+) release (CICR). The cAMP-dependent increase of [Ca(2+)](i) that accompanies CICR is shown to be coupled to exocytosis. We propose that the interaction of cAMP and Epac to trigger CICR explains, at least in part, the blood glucose-lowering properties of an insulinotropic hormone (glucagon-like peptide-1, also known as GLP-1) now under investigation for use in the treatment of type-2 diabetes mellitus.
- Published
- 2003
- Full Text
- View/download PDF
44. Characterisation of PDZ-GEFs, a family of guanine nucleotide exchange factors specific for Rap1 and Rap2
- Author
-
Johannes L. Bos, Johan de Rooij, Fried J. T. Zwartkruis, Holger Rehmann, Alfred Wittinghofer, Miranda van Triest, and H. Bea Kuiperij
- Subjects
Molecular Sequence Data ,PDZ domain ,Nerve Tissue Proteins ,GTPase ,Mice ,cAMP ,Cyclic AMP ,Animals ,Guanine Nucleotide Exchange Factors ,Nucleotide ,splice ,Amino Acid Sequence ,Cloning, Molecular ,Molecular Biology ,chemistry.chemical_classification ,rap1 GTP-Binding Proteins ,3T3 Cells ,Cell Biology ,Family member ,enzymes and coenzymes (carbohydrates) ,rap GTP-Binding Proteins ,Biochemistry ,chemistry ,Cyclic nucleotide-binding domain ,Rap1 ,Guanine nucleotide exchange factor ,sense organs ,Rap ,Ras - Abstract
PDZ-GEF1 (RA-GEF/nRapGEP/CNrasGEF) is a guanine nucleotide exchange factor (GEF) characterised by the presence of a PSD-95/DlgA/ZO-1 (PDZ) domain, a Ras-association (RA) domain and a region related to a cyclic nucleotide binding domain (RCBD). These domains are in addition to a Ras exchange motif (REM) and GEF domain characteristic for GEFs for Ras-like small GTPases. PDZ-GEF1 efficiently exchanges nucleotides of both Rap1 and Rap2, but has also been implicated in mediating cAMP-induced Ras activation through binding of cAMP to the RCBD. Here we describe a new family member, PDZ-GEF2, of which we isolated two splice variants (PDZ-GEF2A and 2B). PDZ-GEF2 contains, in addition to the domains characteristic for PDZ-GEF1, a second, less conserved RCBD at the N-terminus. PDZ-GEF2 is also specific for both Rap1 and Rap2. We further investigated the possibility that PDZ-GEF2, like PDZ-GEF1, is a cAMP-responsive GEF for Ras. However, in contrast to previous results, we did not find any effect of either PDZ-GEF1 or PDZ-GEF2 on Ras in the absence or presence of cAMP. Moreover, affinity measurements by isothermic calorimetry showed that the RCBD of PDZ-GEF1 does not bind cAMP with a physiologically relevant affinity. We conclude that both PDZ-GEF1 and 2 are specific for Rap1 and Rap2 and unresponsive to cAMP and various other nucleotides.
- Published
- 2003
- Full Text
- View/download PDF
45. Cyclic AMP induces integrin-mediated cell adhesion through Epac and Rap1 upon stimulation of the β2-adrenergic receptor
- Author
-
Jorrit M. Enserink, Leo S. Price, Johannes L. Bos, H. Bea Kuiperij, Johan de Rooij, Savithri Rangarajan, and Frank Schwede
- Subjects
Integrins ,Integrin ,8-Bromo Cyclic Adenosine Monophosphate ,integrins ,cyclic nucleotides ,GTPases ,guanine nucleotide exchange factor ,cell adhesion ,Report ,Cyclic AMP Response Element-Binding Protein ,Cell Adhesion ,Cyclic AMP ,Tumor Cells, Cultured ,Guanine Nucleotide Exchange Factors ,Humans ,Cell adhesion ,Protein kinase A ,Ovarian Neoplasms ,biology ,Carcinoma ,Isoproterenol ,rap1 GTP-Binding Proteins ,Cell Biology ,Adrenergic beta-Agonists ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,Fibronectins ,Fibronectin ,biology.protein ,Rap1 ,Female ,Guanine nucleotide exchange factor ,Receptors, Adrenergic, beta-2 ,Signal transduction ,Mitogen-Activated Protein Kinases ,Signal Transduction - Abstract
cAMP controls many cellular processes mainly through the activation of protein kinase A (PKA). However, more recently PKA-independent pathways have been established through the exchange protein directly activated by cAMP (Epac), a guanine nucleotide exchange factor for the small GTPases Rap1 and Rap2. In this report, we show that cAMP can induce integrin-mediated cell adhesion through Epac and Rap1. Indeed, when Ovcar3 cells were treated with cAMP, cells adhered more rapidly to fibronectin. This cAMP effect was insensitive to the PKA inhibitor H-89. A similar increase was observed when the cells were transfected with Epac. Both the cAMP effect and the Epac effect on cell adhesion were abolished by the expression of Rap1–GTPase-activating protein, indicating the involvement of Rap1 in the signaling pathway. Importantly, a recently characterized cAMP analogue, 8-(4-chloro-phenylthio)-2′-O-methyladenosine-3′,5′-cyclic monophosphate, which specifically activates Epac but not PKA, induced Rap-dependent cell adhesion. Finally, we demonstrate that external stimuli of cAMP signaling, i.e., isoproterenol, which activates the Gαs-coupled β2-adrenergic receptor can induce integrin-mediated cell adhesion through the Epac-Rap1 pathway. From these results we conclude that cAMP mediates receptor-induced integrin-mediated cell adhesion to fibronectin through the Epac-Rap1 signaling pathway.
- Published
- 2003
46. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress
- Author
-
Tobias B. Dansen, Karel W. A. Wirtz, Boudewijn M.T. Burgering, Johannes L. Bos, Paul J. Coffer, Ingrid Saarloos, Geert J. P. L. Kops, Paulien E. Polderman, Ting-T. Huang, and René H. Medema
- Subjects
Cell Survival ,Longevity ,Apoptosis ,Protein Serine-Threonine Kinases ,Biology ,medicine.disease_cause ,Models, Biological ,Antioxidants ,Culture Media, Serum-Free ,Superoxide dismutase ,Forkhead Transcription Factors ,Proto-Oncogene Proteins ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Insulin ,RNA, Messenger ,Promoter Regions, Genetic ,chemistry.chemical_classification ,Reactive oxygen species ,Multidisciplinary ,Forkhead Box Protein O1 ,Superoxide Dismutase ,JNK Mitogen-Activated Protein Kinases ,FOXO Family ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Enzyme Activation ,Oxidative Stress ,Glucose ,chemistry ,Biochemistry ,Enzyme Induction ,Mutation ,FOXO4 ,FOXO3 ,biology.protein ,Forkhead Box Protein O3 ,Mitogen-Activated Protein Kinases ,Reactive Oxygen Species ,Proto-Oncogene Proteins c-akt ,Oxidative stress ,Transcription Factors - Abstract
Reactive oxygen species are required for cell proliferation but can also induce apoptosis1. In proliferating cells this paradox is solved by the activation of protein kinase B (PKB; also known as c-Akt), which protects cells from apoptosis2. By contrast, it is unknown how quiescent cells that lack PKB activity are protected against cell death induced by reactive oxygen species. Here we show that the PKB-regulated Forkhead transcription factor FOXO3a (also known as FKHR-L1) protects quiescent cells from oxidative stress by directly increasing their quantities of manganese superoxide dismutase (MnSOD) messenger RNA and protein. This increase in protection from reactive oxygen species antagonizes apoptosis caused by glucose deprivation. In quiescent cells that lack the protective mechanism of PKB-mediated signalling, an alternative mechanism is induced as a consequence of PKB inactivity. This mechanism entails the activation of Forkhead transcription factors, the transcriptional activation of MnSOD and the subsequent reduction of reactive oxygen species. Increased resistance to oxidative stress is associated with longevity. The model of Forkhead involvement in regulating longevity stems from genetic analysis in Caenorhabditis elegans3,4,5,6, and we conclude that this model also extends to mammalian systems.
- Published
- 2002
- Full Text
- View/download PDF
47. The PDZ Domain of the Guanine Nucleotide Exchange Factor PDZGEF Directs Binding to Phosphatidic Acid during Brush Border Formation
- Author
-
Patricia M. Brouwer, Sarah V. Consonni, Eleonora S. van Slobbe, and Johannes L. Bos
- Subjects
PDZ domain ,lcsh:Medicine ,Phosphatidic Acids ,Nerve Tissue Proteins ,chemistry.chemical_compound ,Ezrin ,Intestinal mucosa ,Cell Signaling ,Cell polarity ,Molecular Cell Biology ,Guanine Nucleotide Exchange Factors ,Humans ,Intestinal Mucosa ,lcsh:Science ,Epithelial polarity ,Multidisciplinary ,Microvilli ,lcsh:R ,Mechanisms of Signal Transduction ,Cell Membrane ,Biology and Life Sciences ,Cell Polarity ,Phosphatidic acid ,Cell Biology ,Apical membrane ,Cell biology ,HEK293 Cells ,rap GTP-Binding Proteins ,chemistry ,lcsh:Q ,Guanine nucleotide exchange factor ,Research Article ,Signal Transduction - Abstract
PDZGEF is a guanine nucleotide exchange factor for the small G protein Rap. It was recently found that PDZGEF contributes to establishment of intestinal epithelial polarity downstream of the kinase Lkb1. By binding to phosphatidic acid enriched at the apical membrane, PDZGEF locally activates Rap2a resulting in induction of brush border formation via a pathway that includes the polarity players TNIK, Mst4 and Ezrin. Here we show that the PDZ domain of PDZGEF is essential and sufficient for targeting PDZGEF to the apical membrane of polarized intestinal epithelial cells. Inhibition of PLD and consequently production of phosphatidic acid inhibitis targeting of PDZGEF to the plasma membrane. Furthermore, localization requires specific positively charged residues within the PDZ domain. We conclude that local accumulation of PDZGEF at the apical membrane during establishment of epithelial polarity is mediated by electrostatic interactions between positively charged side chains in the PDZ domain and negatively charged phosphatidic acid.
- Published
- 2014
48. Rap1 signaling in endothelial barrier control
- Author
-
Johannes L. Bos, Willem-Jan Pannekoek, and Anneke Post
- Subjects
rho GTP-Binding Proteins ,endocrine system ,Telomere-Binding Proteins ,CDC42 ,Review ,Biology ,Shelterin Complex ,Cellular and Molecular Neuroscience ,Proto-Oncogene Proteins ,Stress Fibers ,Cell Adhesion ,Guanine Nucleotide Exchange Factors ,Humans ,Cytoskeleton ,cdc42 GTP-Binding Protein ,KRIT1 Protein ,Actin ,rasip1 ,Intracellular Signaling Peptides and Proteins ,cdc42 ,Endothelial Cells ,endothelial barrier function ,Cell Biology ,epac1 ,Actin cytoskeleton ,radil ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Actin Cytoskeleton ,krit1 ,Cdc42 GTP-Binding Protein ,rho ,Rap1 ,Guanine nucleotide exchange factor ,cytoskeletal tension ,Signal transduction ,Microtubule-Associated Proteins ,rap1 ,Signal Transduction - Abstract
The small G-protein Rap1 plays an important role in the regulation of endothelial barrier function, a process controlled largely by cellâ€"cell adhesions and their connection to the actin cytoskeleton. During the various stages of barrier dynamics, different guanine nucleotide exchange factors (GEFs) control Rap1 activity, indicating that Rap1 integrates multiple input signals. Once activated, Rap1 induces numerous signaling cascades, together responsible for the increased endothelial barrier function. Most notably, Rap1 activation results in the inhibition of Rho to decrease radial stress fibers and the activation of Cdc42 to increase junctional actin. This implies that Rap regulates endothelial barrier function by dual control of cytoskeletal tension. The molecular details of the signaling pathways are becoming to be elucidated.
- Published
- 2014
49. Regulation of the Forkhead Transcription Factor AFX by Ral-Dependent Phosphorylation of Threonines 447 and 451
- Author
-
Boudewijn M.T. Burgering, Johannes L. Bos, and Nancy D. de Ruiter
- Subjects
Threonine ,animal structures ,Molecular Sequence Data ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Biology ,Transfection ,Mice ,Structure-Activity Relationship ,Genes, Reporter ,Mutant protein ,Proto-Oncogene Proteins ,Tumor Cells, Cultured ,Animals ,Humans ,Amino Acid Sequence ,Phosphorylation ,Cell Growth and Development ,Molecular Biology ,Protein kinase B ,Transcription factor ,Cell Nucleus ,Regulation of gene expression ,Binding Sites ,Forkhead Transcription Factors ,3T3 Cells ,Cell Biology ,Cell cycle ,Molecular biology ,Enzyme Activation ,Amino Acid Substitution ,Gene Expression Regulation ,Ral GTP-Binding Proteins ,Mutagenesis, Site-Directed ,ras Proteins ,ral GTP-Binding Proteins ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,Transcription Factors - Abstract
AFX is a Forkhead transcription factor that induces a G(1) cell cycle arrest via upregulation of the cell cycle inhibitor p27(Kip1). Previously we have shown that protein kinase B (PKB) phosphorylates AFX causing inhibition of AFX by nuclear exclusion. In addition, Ras, through the activation of the RalGEF-Ral pathway, induces phosphorylation of AFX. Here we show that the Ras-Ral pathway provokes phosphorylation of threonines 447 and 451 in the C terminus of AFX. A mutant protein in which both threonines are substituted for alanines (T447A/T451A) still responds to PKB-regulated nuclear-cytoplasmic shuttling, but transcriptional activity and consequent G(1) cell cycle arrest are greatly impaired. Furthermore, inhibition of the Ral signaling pathway abolishes both AFX-mediated transcription and regulation of p27(Kip1), while activation of Ral augments AFX activity. From these results we conclude that Ral-mediated phosphorylation of threonines 447 and 451 is required for proper activity of AFX-WT. Interestingly, the T447A/T451A mutation did not affect the induction of transcription and G(1) cell cycle arrest by the PKB-insensitive AFX-A3 mutant, suggesting that Ral-mediated phosphorylation plays a role in the regulation of AFX by PKB.
- Published
- 2001
- Full Text
- View/download PDF
50. GTK, a Src-related Tyrosine Kinase, Induces Nerve Growth Factor-independent Neurite Outgrowth in PC12 Cells through Activation of the Rap1 Pathway
- Author
-
Johannes L. Bos, Michael Welsh, Cecilia Annerén, and Kris A. Reedquist
- Subjects
MAPK/ERK pathway ,Neurite ,Cellular differentiation ,Tyrosine phosphorylation ,Cell Biology ,Biology ,Biochemistry ,Cell biology ,Focal adhesion ,chemistry.chemical_compound ,chemistry ,Signal transduction ,Molecular Biology ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The rat pheochromocytoma cell line PC12 is extensively used as a model for studies of neuronal cell differentiation. These cells develop a sympathetic neuron-like phenotype when cultured in the presence of nerve growth factor. The present study was performed in order to assess the role of mouse GTK (previously named BSK/IYK), a cytoplasmic tyrosine kinase belonging to the Src family, for neurite outgrowth in PC12 cells. We report that PC12 cells stably overexpressing GTK exhibit a larger fraction of cells with neurites as compared with control cells, and this response is not accompanied by an increased ERK activity. Treatment of the cells with the MEK inhibitor PD98059 did not reduce the GTK-dependent increased in neurite outgrowth. GTK expression induces a nerve growth factor-independent Rap1 activation, probably through altered CrkII signaling. We observe increased CrkII complex formation with p130Cas, focal adhesion kinase (FAK), and Shb in PC12-GTK cells. The expression of GTK also correlates with a markedly increased content of FAK, phosphorylation of the adaptor protein Shb, and an association between these two proteins. Transient transfection of GTK-overexpressing cells with RalGDS-RBD or Rap1GAP, inhibitors of the Rap1 pathway, reduces the GTK-dependent neurite outgrowth. These data suggest that GTK participates in a signaling pathway, perhaps involving Shb, FAK and Rap1, that induces neurite outgrowth in PC12 cells.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.