Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cell Death Discov ; 9(1): 183, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37321991

ABSTRACT

Uveal melanoma (UM) has a high risk to progress to metastatic disease with a median survival of 3.9 months after metastases detection, as metastatic UM responds poorly to conventional and targeted chemotherapy and is largely refractory to immunotherapy. Here, we present a patient-derived zebrafish UM xenograft model mimicking metastatic UM. Cells isolated from Xmm66 spheroids derived from metastatic UM patient material were injected into 2 days-old zebrafish larvae resulting in micro-metastases in the liver and caudal hematopoietic tissue. Metastasis formation could be reduced by navitoclax and more efficiently by the combinations navitoclax/everolimus and flavopiridol/quisinostat. We obtained spheroid cultures from 14 metastatic and 10 primary UM tissues, which were used for xenografts with a success rate of 100%. Importantly, the ferroptosis-related genes GPX4 and SLC7A11 are negatively correlated with the survival of UM patients (TCGA: n = 80; Leiden University Medical Centre cohort: n = 64), ferroptosis susceptibility is correlated with loss of BAP1, one of the key prognosticators for metastatic UM, and ferroptosis induction greatly reduced metastasis formation in the UM xenograft model. Collectively, we have established a patient-derived animal model for metastatic UM and identified ferroptosis induction as a possible therapeutic strategy for the treatment of UM patients.

2.
Database (Oxford) ; 20222022 04 28.
Article in English | MEDLINE | ID: mdl-35482537

ABSTRACT

Zebrafish xenografts are an established model in cancer biology, with a steadily rising number of models and users. However, as of yet, there is no platform dedicated to standardizing protocols and sharing data regarding zebrafish xenograft phenotypes. Here, we present the Xenograft Phenotype Interactive Repository (XePhIR, https://www.xephir.org) as an independent data-sharing platform to deposit, share and repurpose zebrafish xenograft data. Deposition of data and publication with XePhIR will be done after the acceptation of the original publication. This will enhance the reach of the original research article, enhance visibility and do not interfere with the publication or copyrights of the original article. With XePhIR, we strive to fulfill these objectives and reason that this resource will enhance reproducibility and showcase the appeal and applicability of the zebrafish xenograft model. Database URL: https://www.xephir.org.


Subject(s)
Zebrafish , Animals , Databases, Factual , Heterografts , Humans , Phenotype , Reproducibility of Results , Zebrafish/genetics
3.
Sci Rep ; 9(1): 2399, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30787324

ABSTRACT

Developing tumors interact with the surrounding microenvironment. Myeloid cells exert both anti- and pro-tumor functions and chemokines are known to drive immune cell migration towards cancer cells. It is documented that CXCR4 signaling supports tumor metastasis formation in tissues where CXCL12, its cognate ligand, is abundant. On the other hand, the role of the neutrophilic CXCR4 signaling in driving cancer invasion and metastasis formation is poorly understood. Here, we use the zebrafish xenotransplantation model to study the role of CXCR4 signaling in driving the interaction between invasive human tumor cells and host neutrophils, supporting early metastasis formation. We found that zebrafish cxcr4 (cxcr4b) is highly expressed in neutrophils and experimental micrometastases fail to form in mutant larvae lacking a functional Cxcr4b. We demonstrated that Cxcr4b controls neutrophil number and motility and showed that Cxcr4b transcriptomic signature relates to motility and adhesion regulation in neutrophils in tumor-naïve larvae. Finally, Cxcr4b deficient neutrophils failed to interact with cancer cells initiating early metastatic events. In conclusion, we propose that CXCR4 signaling supports the interaction between tumor cells and host neutrophils in developing tumor metastases. Therefore, targeting CXCR4 on tumor cells and neutrophils could serve as a double bladed razor to limit cancer progression.


Subject(s)
Chemokine CXCL12/genetics , Neoplasms/genetics , Receptors, CXCR4/genetics , Zebrafish Proteins/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Larva/genetics , Mice , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Metastasis , Neoplasms/pathology , Neutrophils/metabolism , Neutrophils/pathology , Transplantation, Heterologous , Tumor Microenvironment/genetics , Zebrafish/genetics
4.
Adv Exp Med Biol ; 916: 239-63, 2016.
Article in English | MEDLINE | ID: mdl-27165357

ABSTRACT

Tumor angiogenesis and metastasis are key steps of cancer progression. In vitro and animal model studies have contributed to partially elucidating the mechanisms involved in these processes and in developing therapies. Besides the improvements in fundamental research and the optimization of therapeutic regimes, cancer still remains a major health threatening condition and therefore the development of new models is needed. The zebrafish is a powerful tool to study tumor angiogenesis and metastasis, because it allows the visualization of fluorescently labelled tumor cells inducing vessel remodeling, disseminating and invading surrounding tissues in a whole transparent embryo. The embryo model has also been used to address the contribution of the tumor stroma in sustaining tumor angiogenesis and spreading. Simultaneously, new anti-angiogenic drugs and compounds affecting malignant cell survival and migration can be tested by simply adding the compound into the water of living embryos. Therefore the zebrafish model offers the opportunity to gain more knowledge on cancer angiogenesis and metastasis in vivo with the final aim of providing new translational insights into therapeutic approaches to help patients.


Subject(s)
Disease Models, Animal , Neoplasms/blood supply , Neovascularization, Pathologic , Zebrafish/embryology , Animals , Fluorescent Dyes , Heterografts , Neoplasm Metastasis , Neoplasms/pathology
5.
Eur J Cancer ; 51(17): 2624-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26428435

ABSTRACT

PURPOSE: Ewing sarcoma (EWS) is the second most common sarcoma of bone in children and young adults. Patients with disseminated disease at diagnosis or early relapse have a poor prognosis. Our goal was to identify novel predictive biomarkers for these patients, focusing on chemokines, specifically genes involved in the CXCR4-pathway because of their established role in metastasis and tumour growth. METHODS: Total RNA isolated from therapy-naïve tumour samples (n=18; panel I) and cell lines (n=21) was used to study expression of CXCR4-pathway related genes and CXCR4 splice variants (CXCR4-2: Small and CXCR4-1: Large) by RT-Q-PCR. Expression levels were correlated to overall survival (OS) and event free survival (EFS). Study results were validated in an independent series of 26 tumour samples (panel II) from therapy-naïve tumour samples. RESULTS: CXCL12, CXCR4, CXCR7 and CXCL14 were expressed and high CXCR7 and CXCL14 expression showed a positive correlation with EFS and OS and a negative correlation with metastasis development. Both splice variants CXCR4 were expressed in cell lines and tumour samples and CXCR4-1/CXCR4-2 ratio was significantly higher in tumour samples compared to cell lines and correlated with an improved EFS and OS. The results from the test panel were validated in an independent sample panel. CONCLUSIONS: We identified a set of genes involved in CXCR4 signalling that may be used as a marker to predict survival and metastasis development in Ewing sarcoma.


Subject(s)
Bone Neoplasms/genetics , Chemokines, CXC/genetics , Gene Expression Regulation, Neoplastic , Receptors, CXCR4/genetics , Receptors, CXCR/genetics , Sarcoma, Ewing/genetics , Adolescent , Adult , Bone Neoplasms/pathology , Cell Line, Tumor , Chemokine CXCL12/genetics , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis , Protein Isoforms/genetics , RNA Splicing , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/pathology , Signal Transduction/genetics , Young Adult
6.
Cancer Res ; 75(11): 2326-36, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25858144

ABSTRACT

Altered microRNA (miRNA; miR) expression is associated with tumor formation and progression of various solid cancers. A major challenge in miRNA expression profiling of bulk tumors is represented by the heterogeneity of the subpopulations of cells that constitute the organ, as well as the tumor tissue. Here, we analyzed the expression of miRNAs in a subpopulation of epithelial stem/progenitor-like cells in human prostate cancer [prostate cancer stem cell (PCSC)] and compared their expression profile to more differentiated cancer cells. In both cell lines and clinical prostate cancer specimens, we identified that miR-25 expression in PCSCs was low/absent and steadily increased during their differentiation into cells with a luminal epithelial phenotype. Functional studies revealed that overexpression of miR-25 in prostate cancer cell lines and selected subpopulation of highly metastatic and tumorigenic cells (ALDH(high)) strongly affected the invasive cytoskeleton, causing reduced migration in vitro and metastasis via attenuation of extravasation in vivo. Here, we show, for the first time, that miR-25 can act as a tumor suppressor in highly metastatic PCSCs by direct functional interaction with the 3'-untranslated regions of proinvasive αv- and α6-integrins. Taken together, our observations suggest that miR-25 is a key regulator of invasiveness in human prostate cancer through its direct interactions with αv- and α6-integrin expression.


Subject(s)
Integrin alpha6/biosynthesis , Integrin alphaV/biosynthesis , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic/genetics , Humans , Integrin alpha6/genetics , Integrin alphaV/genetics , Male , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology
7.
Methods Enzymol ; 521: 47-67, 2013.
Article in English | MEDLINE | ID: mdl-23351733

ABSTRACT

The lateral diffusion of a G-protein-coupled receptor (GPCR) in the plasma membrane determines its interaction capabilities with downstream signaling molecules and critically modulates its function. Mechanisms that control GPCR mobility, like compartmentalization, enable a cell to fine-tune its response through local changes in the rate, duration, and extent of signaling. These processes are known to be highly dynamic and tightly regulated in time and space, usually not completely synchronized in time. Therefore, bulk studies such as protein biochemistry or conventional confocal microscopy will only yield information on the average properties of the interactions and are compromised by poor time resolution. Single-particle tracking (SPT) in living cells is a key approach to directly monitor the function of a GPCR within its natural environment and to obtain unprecedented detailed information about receptor mobility, binding kinetics, aggregation states, and domain formation. This review provides a detailed description on how to perform single GPCR tracking experiments.


Subject(s)
Cell Membrane/metabolism , Molecular Imaging/methods , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/metabolism , Cell Membrane/chemistry , Dictyostelium/cytology , Dictyostelium/metabolism , Equipment Design , Humans , Models, Molecular , Molecular Imaging/instrumentation , Protozoan Proteins/analysis , Protozoan Proteins/metabolism
8.
Oncogene ; 31(39): 4333-42, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22179836

ABSTRACT

Angiogenesis is essential for development and tumor progression. With the aim of identifying new compound inhibitors of the angiogenesis process, we used an established enhanced green fluorescent protein-transgenic zebrafish line to develop an automated assay that enables high-throughput screening of compound libraries in a whole-organism setting. Using this system, we have identified novel kinase inhibitor compounds that show anti-angiogenic properties in both zebrafish in-vivo system and in human endothelial cell in-vitro angiogenesis models. Furthermore, we have determined the kinase target of these compounds and have identified and validated a previously uncharacterized involvement of phosphorylase kinase subunit G1 (PhKG1) in angiogenesis in vivo. In addition, we have found that PhKG1 is upregulated in human tumor samples and that aberrations in gene copy number of PhK subunits are a common feature of human tumors. Our results provide a novel insight into the angiogenesis process, as well as identify new potential targets for anti-angiogenic therapies.


Subject(s)
Angiogenesis Inhibitors/isolation & purification , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Phosphorylase Kinase/antagonists & inhibitors , Zebrafish , Angiogenesis Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Cell Line , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Gene Dosage , High-Throughput Screening Assays , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Phosphorylase Kinase/genetics , Up-Regulation
9.
Biochem Biophys Res Commun ; 282(3): 765-72, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11401529

ABSTRACT

Exogenous lysophosphatidic acid (LPA) has been shown to evoke a chemotactic response in aggregative cells of the social amoeba Dictyostelium discoideum. In this paper, we demonstrate that extracellular LPA is also able to induce activation of mitogen-activated protein (MAP) kinase DdERK2 (extracellular signal regulated kinase 2) in these cells. This activation is independent of cyclic AMP receptors, yet fully dependent on the single Gbeta subunit, hinting to the presence of functional heptahelical LPA receptors in a primitive eukaryote. We did not observe LPA-dependent cyclic GMP accumulation, which suggests that the pathways for LPA-induced and "classical" chemotaxis of D. discoideum cells are substantially different.


Subject(s)
Dictyostelium/physiology , Heterotrimeric GTP-Binding Proteins/metabolism , Lysophospholipids/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Receptors, G-Protein-Coupled , Animals , Chemotaxis/drug effects , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dictyostelium/drug effects , Dictyostelium/genetics , Enzyme Activation/drug effects , Lysophospholipids/metabolism , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/metabolism , Receptors, Lysophosphatidic Acid , Signal Transduction
10.
Biochem Biophys Res Commun ; 260(2): 504-9, 1999 Jul 05.
Article in English | MEDLINE | ID: mdl-10403797

ABSTRACT

Ca(2+) influx and mitogen-activated protein (MAP) kinase activation are important phenomena in signal transduction, which are often interconnected. We investigated whether serpentine receptor-dependent, Gbeta-independent activation of MAP kinase ERK2 by chemoattractant cyclic AMP (cAMP) is mediated by Ca(2+) influx in the social amoeba Dictyostelium discoideum. We generated a D. discoideum double mutant, which harbours a temperature-sensitive Gbeta subunit and expresses the apoaequorin protein. Utilizing this mutant, we demonstrate that cAMP induced Ca(2+) influx into intact D. discoideum cells can be blocked completely at both the permissive and the restrictive temperature, by using either gadolinium ions or Ruthenium Red. Under the same experimental conditions, these substances do not abolish cAMP stimulation of ERK2 at either temperature. We conclude that there is a Gbeta- and Ca(2+) influx-independent pathway for the receptor-dependent activation of MAP kinase ERK2 in D. discoideum.


Subject(s)
Calcium/metabolism , Dictyostelium/enzymology , GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Receptors, Peptide/metabolism , Transcription Factors , Animals , Cyclic AMP/metabolism , Enzyme Activation , Ion Transport , Receptors, Mating Factor
11.
Biochim Biophys Acta ; 1449(1): 1-24, 1999 Feb 04.
Article in English | MEDLINE | ID: mdl-10076047

ABSTRACT

Cells can react to environmental changes by transduction of extracellular signals, to produce intracellular responses. Membrane-impermeable signal molecules are recognized by receptors, which are localized on the plasma membrane of the cell. Binding of a ligand can result in the stimulation of an intrinsic enzymatic activity of its receptor or the modulation of a transducing protein. The modulation of one or more intracellular transducing proteins can finally lead to the activation or inhibition of a so-called 'effector protein'. In many instances, this also results in altered gene expression. Phosphorylation by protein kinases is one of the most common and important regulatory mechanisms in signal transmission. This review discusses the non-channel transmembrane receptors and their downstream signaling, with special focus on the role of protein kinases.


Subject(s)
Protein Kinases/physiology , Receptors, Cell Surface/physiology , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Humans , Phosphorylation , Plants , Protein Kinase C/physiology , Receptor Protein-Tyrosine Kinases/physiology , Saccharomyces
12.
Biochim Biophys Acta ; 1357(2): 243-8, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9223628

ABSTRACT

A 1.0 kb cDNA clone (Dd14-3-3) encoding a 14-3-3 homologue was isolated from a Dictyostelium discoideum cDNA library. The putative Dd14-3-3 protein has highest sequence identity to a barley 14-3-3 isoform (74%). Southern blot analysis suggests that only one 14-3-3 gene is present in the Dictyostelium genome. Highest Dd14-3-3 expression is observed in vegetatively growing cells, and expression decreases during multicellular development. In contrast, Dd14-3-3 protein levels detected immunochemically remained constant during Dictyostelium development. Expression of the Dd14-3-3 cDNA in Saccharomyces cerevisiae complemented the lethal disruption of the two yeast genes encoding 14-3-3 proteins (BMH1 and BMH2). This shows that Dd14-3-3 can fulfil the same function(s) as the yeast 14-3-3 proteins.


Subject(s)
Dictyostelium/genetics , Proteins/genetics , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , Blotting, Western , DNA, Complementary/chemistry , DNA, Complementary/isolation & purification , Dictyostelium/growth & development , Gene Expression , Hordeum/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Proteins/chemistry , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Sequence Homology
13.
EMBO J ; 15(13): 3361-8, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8670837

ABSTRACT

Dictyostelium discoideum expresses two Extracellular signal Regulated Kinases, ERK1 and ERK2, which are involved in growth, multicellular development and regulation of adenylyl cyclase. Binding of extracellular cAMP to cAMP receptor 1, a G-protein coupled cell surface receptor, transiently stimulates phosphorylation, activation and nuclear translocation of ERK2. Activation of ERK2 by cAMP is dependent on heterotrimeric G-proteins, since activation of ERK2 is absent in cells lacking the Galpha4 subunit. The small G-protein rasD also activates ERK2. In cells overexpressing a mutated, constitutively active rasD, ERK2 activity is elevated prior to cAMP stimulation. Intracellular cAMP and cAMP-dependent protein kinase (PKA) are essential for adaptation of the ERK2 response. This report shows that multiple signalling pathways are involved in regulation of ERK2 activity in D.discoideum.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP/physiology , Dictyostelium/enzymology , GTP-Binding Proteins/physiology , ras Proteins/physiology , Animals , Biological Transport , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Nucleus/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Mitogen-Activated Protein Kinase 1 , Phosphorylation , Receptors, Cell Surface/metabolism
14.
Plant Cell ; 8(6): 1061-1067, 1996 Jun.
Article in English | MEDLINE | ID: mdl-12239411

ABSTRACT

Abscisic acid (ABA) induces a rapid and transient mitogen-activated protein (MAP) kinase activation in barley aleurone protoplasts. MAP kinase activity, measured as myelin basic protein phosphorylation by MAP kinase immunoprecipitates, increased after 1 min, peaked after 3 min, and decreased to basal levels after ~5 min of ABA treatment in vivo. Antibodies recognizing phosphorylated tyrosine residues precipitate with myelin basic protein kinase activity that has identical ABA activation characteristics and demonstrate that tyrosine phosphorylation of MAP kinase occurs during activation. The half-maximal concentration of ABA required for MAP kinase activation, 3 x 10-7 M, is very similar to that required for ABA-induced rab16 gene expression. The tyrosine phosphatase inhibitor phenylarsine oxide can completely block ABA-induced MAP kinase activation and rab16 gene expression. These results lead us to conclude that ABA activates MAP kinase via a tyrosine phosphatase and that these steps are a prerequisite for ABA induction of rab16 gene expression.

16.
FEBS Lett ; 329(3): 245-8, 1993 Aug 30.
Article in English | MEDLINE | ID: mdl-8365465

ABSTRACT

We report the existence of several families of GTP-binding proteins in barley aleurone protoplasts. Partial purified plasma membrane proteins were separated by SDS-PAGE, transferred to a nitrocellulose filter and incubated with either antisera raised against a highly conserved animal G protein alpha subunit peptide/or Ras protein, or with [alpha-32P]GTP. Two sets of proteins of M(r) = 32-36 kDa and 22-24 kDa were strongly recognized by the antisera. Binding of [alpha-32P]GTP was detected on Western blots with proteins of M(r) = 22-24 kDa and 16 kDa. Binding was inhibited by 10(-7)-10(-6) M GTP gamma S, GTP or GDP; binding was not affected by 10(-6)-10(-5) M ATP gamma S or ADP. The kinetics, specificity and the effects of phytohormones in a [35S]GTP gamma S binding assay were also studied in isolated plasma membranes of barley aleurone protoplasts.


Subject(s)
GTP-Binding Proteins/analysis , Hordeum/chemistry , Protoplasts/chemistry , Binding Sites , Blotting, Western , Cell Membrane/metabolism , Electrophoresis, Polyacrylamide Gel , GTP-Binding Proteins/metabolism , Plant Growth Regulators/pharmacology , Signal Transduction , Temperature
17.
Eur J Biochem ; 209(1): 299-304, 1992 Oct 01.
Article in English | MEDLINE | ID: mdl-1356770

ABSTRACT

Li+ drastically alters pattern formation in Dictyostelium by inhibiting cAMP-induced prespore-gene expression and promoting cAMP-induced prestalk-gene expression. We reported previously that Li+ inhibits inositol monophosphatases in this organism and strongly reduces basal and cAMP-stimulated inositol 1,4,5-trisphosphate levels. We show here that Li+ also reduces cAMP-induced accumulation of cAMP, but promotes cAMP-induced accumulation of cGMP. This effect is not due to inhibition of cGMP hydrolysis or inhibition of adaptation and may therefore reflect stimulation of guanylyl-cyclase activation. Li+ does not affect the binding of cAMP to surface receptors but interferes with the interaction between receptors and guanine-nucleotide-binding regulatory (G) proteins. These effects are complex; in the absence of Mg2+, Li+ increases guanosine 5'-[gamma-thio]triphosphate(GTP[S])-binding activity to similar levels as 1 mM Mg2+. However, while Mg2+ potentiates cAMP-induced stimulation of GTP[S]-binding activity, Li+ effectively inhibits stimulation. Li+ also inhibits cAMP-stimulated, but not basal high-affinity GTP-ase activity, indicating an inhibitory effect on cAMP-induced activation of G-proteins. Our data suggest that in addition to inositolphosphate metabolism, the activation of G-proteins may be a second biochemical target for Li+ effects on pattern formation and signal transduction in Dictyostelium.


Subject(s)
Adenylyl Cyclases/metabolism , Cyclic AMP/pharmacology , Dictyostelium/metabolism , GTP-Binding Proteins/antagonists & inhibitors , Guanylate Cyclase/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Lithium/pharmacology , Animals , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dictyostelium/drug effects , Enzyme Activation/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Second Messenger Systems/drug effects , Signal Transduction/drug effects
18.
Eur J Biochem ; 208(2): 235-40, 1992 Sep 01.
Article in English | MEDLINE | ID: mdl-1325900

ABSTRACT

Drug-resistance selection in Dictyostelium discoideum transformants resulted in up to eight-times-higher ras protein levels. Over-production of the wild-type ras protein did not lead to an aberrant phenotype. Increased levels of the mutated [G12T]ras protein, however, were correlated with severe deficiencies in aggregation and development. This aberrant phenotype is associated with reduced cAMP binding, due to a lower number of cell-surface receptors. We show that both RNA and cAMP-receptor-protein levels are reduced. These results indicate that ras in Dictyostelium discoideum seems to be involved in regulating cAMP-receptor-gene expression.


Subject(s)
Dictyostelium/genetics , Gene Expression Regulation, Fungal , Genes, ras/genetics , Receptors, Cyclic AMP/metabolism , Animals , Blotting, Northern , Blotting, Western , Cyclic AMP/metabolism , Drug Resistance, Microbial/genetics , Gentamicins , Mutagenesis , Phenotype , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Cyclic AMP/genetics , Transformation, Genetic
19.
J Cell Sci ; 100 ( Pt 4): 825-31, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1667662

ABSTRACT

Dictyostelium cells use extracellular cyclic AMP both as a chemoattractant and as a morphogen inducing cell-type-specific gene expression. Cyclic AMP binds to surface receptors, activates one or more G-proteins, and stimulates adenylate cyclase, guanylate cyclase and phosphoinositidase C. Mutant fgdC showed aberrant chemotaxis, and was devoid of cyclic AMP-induced gene expression and differentiation. Both the receptor- and G-protein-mediated stimulation of adenylate cyclase and guanylate cyclase were unaltered in mutant fgdC as compared to wild-type cells. In wild-type cells phosphoinositidase C was activated about twofold by the cyclic AMP receptor. In mutant fgdC cells, however, the enzyme was inhibited by about 60%. These results suggest that phosphoinositidase C is regulated by a receptor-operated activation/inhibition switch that is defective in mutant fgdC. We conclude that activation of phosphoinositidase C is essential for Dictyostelium development.


Subject(s)
Chemotaxis/physiology , Cyclic AMP/pharmacology , Dictyostelium/physiology , Gene Expression Regulation, Enzymologic/drug effects , Phosphoric Diester Hydrolases/metabolism , Animals , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Mutation , Receptors, Cyclic AMP/metabolism , Second Messenger Systems
20.
Proc Natl Acad Sci U S A ; 88(20): 9219-23, 1991 Oct 15.
Article in English | MEDLINE | ID: mdl-11607223

ABSTRACT

During development of the cellular slime mold Dictyostelium discoideum, cAMP induces chemotaxis and expression of different classes of genes by means of interaction with surface cAMP receptors. We describe a cAMP derivative, 8-p-chlorophenylthioadenosine 3',5'-cyclic monophosphate (8-CPT-cAMP), which inhibits cAMP-induced chemotaxis at low concentrations but induces chemotaxis at supersaturating concentrations. This compound, moreover, selectively activates expression of aggregative genes but not of postaggregative genes. 8-CPT-cAMP induces normal cGMP and cAMP accumulation but in contrast to cAMP, which increases inositol 1,4,5-trisphosphate levels, 8-CPT-cAMP decreases inositol 1,4,5-trisphosphate levels. The derivative induces reduced activation of guanine nucleotide regulatory proteins, which may cause its defective activation of inositol 1,4,5-trisphosphate production. Our data suggest that disruption of inositolphospholipid signaling impairs chemotaxis and expression of a subclass of cAMP-regulated genes.

SELECTION OF CITATIONS
SEARCH DETAIL
...