Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Stem Cell Res Ther ; 10(1): 130, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31036072

ABSTRACT

BACKGROUND: Pluripotent stem cells are attractive progenitor cells for the generation of erythroid cells in vitro as have expansive proliferative potential. However, although embryonic (ESC) and induced pluripotent (iPSC) stem cells can be induced to undergo erythroid differentiation, the majority of cells fail to enucleate and the molecular basis of this defect is unknown. One protein that has been associated with the initial phase of erythroid cell enucleation is the intermediate filament vimentin, with loss of vimentin potentially required for the process to proceed. METHODS: In this study, we used our established erythroid culture system along with western blot, PCR and interegation of comparative proteomic data sets to analyse the temporal expression profile of vimentin in erythroid cells differentiated from adult peripheral blood stem cells, iPSC and ESC throughout erythropoiesis. Confocal microscopy was also used to examine the intracellular localisation of vimentin. RESULTS: We show that expression of vimentin is turned off early during normal adult erythroid cell differentiation, with vimentin protein lost by the polychromatic erythroblast stage, just prior to enucleation. In contrast, in erythroid cells differentiated from iPSC and ESC, expression of vimentin persists, with high levels of both mRNA and protein even in orthochromatic erythroblasts. In the vimentin-positive iPSC orthochromatic erythroblasts, F-actin was localized around the cell periphery; however, in those rare cells captured undergoing enucleation, vimentin was absent and F-actin was re-localized to the enucleosome as found in normal adult orthrochromatic erythroblasts. CONCLUSION: As both embryonic and adult erythroid cells loose vimentin and enucleate, retention of vimentin by iPSC and ESC erythroid cells indicates an intrinsic defect. By analogy with avian erythrocytes which naturally retain vimentin and remain nucleated, retention in iPSC- and ESC-derived erythroid cells may impede enucleation. Our data also provide the first evidence that dysregulation of processes in these cells occurs from the early stages of differentiation, facilitating targeting of future studies.


Subject(s)
Erythropoiesis/physiology , Induced Pluripotent Stem Cells/metabolism , Proteomics/methods , Vimentin/metabolism , Cell Differentiation , Cells, Cultured , Erythroid Cells , Humans , Induced Pluripotent Stem Cells/cytology
2.
Nat Commun ; 10(1): 881, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30787325

ABSTRACT

Red blood cells mature within the erythroblastic island (EI) niche that consists of specialized macrophages surrounded by differentiating erythroblasts. Here we establish an in vitro system to model the human EI niche using macrophages that are derived from human induced pluripotent stem cells (iPSCs), and are also genetically programmed to an EI-like phenotype by inducible activation of the transcription factor, KLF1. These EI-like macrophages increase the production of mature, enucleated erythroid cells from umbilical cord blood derived CD34+ haematopoietic progenitor cells and iPSCs; this enhanced production is partially retained even when the contact between progenitor cells and macrophages is inhibited, suggesting that KLF1-induced secreted proteins may be involved in this enhancement. Lastly, we find that the addition of three secreted factors, ANGPTL7, IL-33 and SERPINB2, significantly enhances the production of mature enucleated red blood cells. Our study thus contributes to the ultimate goal of replacing blood transfusion with a manufactured product.


Subject(s)
Erythroblasts/cytology , Erythrocytes/cytology , Erythropoiesis/physiology , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Transcription Factors/metabolism , Macrophages/cytology , Angiopoietin-Like Protein 7 , Angiopoietin-like Proteins/metabolism , Antigens, CD34/metabolism , Blood Substitutes/therapeutic use , Blood Transfusion , Hematopoietic Stem Cells/cytology , Humans , Interleukin-33/metabolism , Kruppel-Like Transcription Factors/genetics , Plasminogen Activator Inhibitor 2/metabolism
3.
Stem Cells Transl Med ; 5(10): 1394-1405, 2016 10.
Article in English | MEDLINE | ID: mdl-27400796

ABSTRACT

: This article describes a good manufacturing practice (GMP)-compatible, feeder-free and serum-free method to produce large numbers of erythroid cells from human pluripotent stem cells (hPSCs), either embryonic or induced. This multistep protocol combines cytokines and small molecules to mimic and surpass the early stages of development. It produces, without any selection or sorting step, a population of cells in which 91.8% ± 5.4% express CD34 at day 7, 98.6% ± 1.3% express CD43 at day 10, and 99.1% ± 0.95% of cells are CD235a positive by day 31 of the differentiation process. Moreover, this differentiation protocol supports extensive expansion, with a single hPSC producing up to 150 hematopoietic progenitor cells by day 10 and 50,000-200,000 erythroid cells by day 31. The erythroid cells produced exhibit a definitive fetal hematopoietic type, with 90%-95% fetal globin and variable proportion of embryonic and adult globin at the protein level. The presence of small molecules during the differentiation protocol has quantitative and qualitative effects; it increases the proportion of adult globin and decreases the proportion of embryonic globin. Given its level of definition, this system provides a powerful tool for investigation of the mechanisms governing early hematopoiesis and erythropoiesis, including globin switching and enucleation. The early stages of the differentiation protocol could also serve as a starting point for the production of endothelial cells and other hematopoietic cells, or to investigate the production of long-term reconstituting hematopoietic stem cells from hPSCs. SIGNIFICANCE: This differentiation protocol allows the production of a large amount of erythroid cells from pluripotent stem cells. Its efficiency is compatible with that of in vitro red blood cell production, and it can be a considerable asset for studying developmental erythropoiesis and red blood cell enucleation, thereby aiding both basic and translational research. In addition to red cells, the early stages of the protocol could also be used as a starting point for the large-scale production of other hematopoietic cell types, including the ultimate goal of generating long-term reconstituting hematopoietic stem cells.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Erythrocytes/cytology , Pluripotent Stem Cells/cytology , Cell Line , Humans
4.
Stem Cells ; 30(4): 643-54, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22232059

ABSTRACT

MicroRNAs (miRNAs) are short noncoding RNAs, which post-transcriptionally regulate gene expression. miRNAs are transcribed as precursors and matured to active forms by a series of enzymes, including Dicer. miRNAs are important in governing cell differentiation, development, and disease. We have recently developed a feeder- and serum-free protocol for direct derivation of endothelial cells (ECs) from human embryonic stem cells (hESCs) and provided evidence of increases in angiogenesis-associated miRNAs (miR-126 and -210) during the process. However, the functional role of miRNAs in hESC differentiation to vascular EC remains to be fully interrogated. Here, we show that the reduction of miRNA maturation induced by Dicer knockdown suppressed hES-EC differentiation. A miRNA microarray was performed to quantify hES-EC miRNA profiles during defined stages of endothelial differentiation. miR-99b, -181a, and -181b were identified as increasing in a time- and differentiation-dependent manner to peak in mature hESC-ECs and adult ECs. Augmentation of miR-99b, -181a, and -181b levels by lentiviral-mediated transfer potentiated the mRNA and protein expression of EC-specific markers, Pecam1 and VE Cadherin, increased nitric oxide production, and improved hES-EC-induced therapeutic neovascularization in vivo. Conversely, knockdown did not impact endothelial differentiation. Our results suggest that miR-99b, -181a, and -181b comprise a component of an endothelial-miRNA signature and are capable of potentiating EC differentiation from pluripotent hESCs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endothelial Cells/cytology , MicroRNAs/genetics , Adult , Biomarkers/metabolism , Cell Line , Cell Lineage/genetics , DEAD-box RNA Helicases/metabolism , Endothelial Cells/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Lentivirus/genetics , MicroRNAs/metabolism , Neovascularization, Physiologic/genetics , Nitric Oxide/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Reproducibility of Results , Ribonuclease III/metabolism , Transcriptome/genetics
5.
Biochem J ; 432(3): 575-84, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20923411

ABSTRACT

hESCs (human embryonic stem cells) have enormous potential for use in pharmaceutical development and therapeutics; however, to realize this potential, there is a requirement for simple and reproducible cell culture methods that provide adequate numbers of cells of suitable quality. We have discovered a novel way of blocking the spontaneous differentiation of hESCs in the absence of exogenous cytokines by supplementing feeder-free conditions with EHNA [erythro-9-(2-hydroxy-3-nonyl)adenine], an established inhibitor of ADA (adenosine deaminase) and cyclic nucleotide PDE2 (phosphodiesterase 2). hESCs maintained in feeder-free conditions with EHNA for more than ten passages showed no reduction in hESC-associated markers including NANOG, POU5F1 (POU domain class 5 transcription factor 1, also known as Oct-4) and SSEA4 (stage-specific embryonic antigen 4) compared with cells maintained in feeder-free conditions containing bFGF (basic fibroblast growth factor). Spontaneous differentiation was reversibly suppressed by the addition of EHNA, but, upon removing EHNA, hESC populations underwent efficient spontaneous, multi-lineage and directed differentiation. EHNA also acts as a strong blocker of directed neuronal differentiation. Chemically distinct inhibitors of ADA and PDE2 lacked the capacity of EHNA to suppress hESC differentiation, suggesting that the effect is not driven by inhibition of either ADA or PDE2. Preliminary structure-activity relationship analysis found the differentiation-blocking properties of EHNA to reside in a pharmacophore comprising a close adenine mimetic with an extended hydrophobic substituent in the 8- or 9-position. We conclude that EHNA and simple 9-alkyladenines can block directed neuronal and spontaneous differentiation in the absence of exogenous cytokine addition, and may provide a useful replacement for bFGF in large-scale or cGMP-compliant processes.


Subject(s)
Adenine/analogs & derivatives , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Adenine/pharmacology , Adenosine Deaminase Inhibitors/pharmacology , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Culture Techniques/methods , Cell Line , Embryonic Stem Cells/cytology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Nanog Homeobox Protein , Neurons/drug effects , Neurons/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphodiesterase Inhibitors/pharmacology , Pluripotent Stem Cells/cytology , Second Messenger Systems/drug effects , Stage-Specific Embryonic Antigens/metabolism , Structure-Activity Relationship , Time Factors
6.
Biochem Soc Trans ; 38(4): 1058-61, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20659003

ABSTRACT

hESCs (human embryonic stem cells) offer great potential for pharmaceutical research and development and, potentially, for therapeutic use. However, improvements in cell culture are urgently required to allow the scalable production of large numbers of cells that maintain pluripotency. Supplementing feeder-free conditions with either EHNA [erythro-9-(2-hydroxy-3-nonyl)adenine] or readily synthesized analogues of this compound maintains hESC pluripotency in the absence of exogenous cytokines. When the hESC lines SA121 or SA461 were maintained in feeder-free conditions with EHNA they displayed no reduction in stem-cell-associated markers such as Nanog, Oct4 (octamer-binding protein 4) and SSEA4 (stage-specific embryonic antigen 4) when compared with cells maintained in full feeder-free conditions that included exogenously added bFGF (basic fibroblast growth factor). Spontaneous differentiation was reversibly suppressed by the addition of EHNA, but EHNA did not limit efficient spontaneous or directed differentiation following its removal. We conclude that EHNA or related compounds offers a viable alternative to exogenous cytokine addition in maintaining hESC cultures in a pluripotent state and might be a particularly useful replacement for bFGF for large-scale or GMP (good manufacturing practice)-compliant processes.


Subject(s)
Embryonic Stem Cells/drug effects , Pluripotent Stem Cells/drug effects , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Cell Culture Techniques , Cell Differentiation/drug effects , Embryonic Stem Cells/physiology , Humans , Ligands , Pluripotent Stem Cells/physiology
7.
Cell Signal ; 20(11): 2071-83, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18721873

ABSTRACT

Transcripts for the PDE4A10 cyclic AMP phosphodiesterase isoform are present in a wide variety of rat tissues including the heart. Sequence comparisons between the putative human and mouse promoters revealed a number of conserved regions including both an Sp1 and a CREB-binding site. The putative mouse PDE4A10 promoter was amplified from genomic DNA and sub-cloned into a luciferase reporter vector for investigation of activity in neonatal cardiac myocytes. Transfection with this construct identified a high level of luciferase expression in neonatal cardiac myocytes. Surprisingly, this activity was down-regulated by elevation of intracellular cAMP through a process involving PKA, but not EPAC, signalling. Such inhibition of the rodent PDE4A10 promoter activity in response to elevated cAMP levels is in contrast to the PDE4 promoters so far described. Site-directed mutagenesis revealed that the Sp1 binding site at promoter position -348 to -336 is responsible for the basal constitutive expression of murine PDE4A10. The conserved CREB-binding motif at position -370 to -363 also contributes to basal promoter activity but does not in itself confer cAMP inhibition upon the PDE4A10 promoter. EMSA analysis confirmed the authenticity of CREB and Sp1 binding sites. The transcriptional start site was identified to be an adenine residue at position -55 in the mouse PDE4A10 promoter. We present evidence that this novel down-regulation of PDE4A10 is mediated by the transcription factor ICER in a PKA dependent manner. The pool of cAMP in cardiac myocytes that down-regulates PDE4A10 is regulated by beta-adrenoceptor coupled adenylyl cyclase activity and via hydrolysis determined predominantly by the action of PDE4 (cAMP phosphodiesterase-4) and not PDE3 (cAMP phosphodiesterase-3). We suggest that increased cAMP may remodel cAMP-mediated signalling events by not only increasing the expression of specific PDE4 cAMP phosphodiesterases but also by down-regulating specific isoforms, such as is shown here for PDE4A10 in cardiac myocytes.


Subject(s)
Cyclic AMP/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Down-Regulation/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Promoter Regions, Genetic , Animals , Animals, Newborn , Base Sequence , Binding Sites , Colforsin/pharmacology , Conserved Sequence , Cyclic AMP/analogs & derivatives , Cyclic AMP Response Element Modulator/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Mutational Analysis , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Humans , Mice , Molecular Sequence Data , Mutation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Response Elements/genetics , Sp1 Transcription Factor/metabolism , Transcription Initiation Site , Transcription, Genetic/drug effects
8.
Cell Signal ; 19(12): 2612-24, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17900862

ABSTRACT

The unique 88 amino acid N-terminal region of cAMP-specific phosphodiesterase-4D5 (PDE4D5) contains overlapping binding sites conferring interaction with the signaling scaffold proteins, betaarrestin and RACK1. A 38-mer peptide, whose sequence reflected residues 12 through 49 of PDE4D5, encompasses the entire N-terminal RACK1 Interaction Domain (RAID1) together with a portion of the beta-arrestin binding site. (1)H NMR and CD analyses indicate that this region has propensity to form a helical structure. The leucine-rich hydrophobic grouping essential for RACK1 interaction forms a discrete hydrophobic ridge located along a single face of an amphipathic alpha-helix with Arg34 and Asn36, which also play important roles in RACK1 binding. The Asn22/Pro23/Trp24/Asn26 grouping, essential for RACK1 interaction, was located at the N-terminal head of the amphipathic helix that contained the hydrophobic ridge. RAID1 is thus provided by a distinct amphipathic helical structure. We suggest that the binding of PDE4D5 to the WD-repeat protein, RACK1, may occur in a manner akin to the helix-helix interaction shown for G(gamma) binding to the WD-repeat protein, G(beta). A more extensive section of the PDE4D5 N-terminal sequence (Thr11-Ala85) is involved in beta-arrestin binding. Several residues within the RAID1 helix contribute to this interaction however. We show here that these residues form a focused band around the centre of the RAID1 helix, generating a hydrophobic patch (from Leu29, Val30 and Leu33) flanked by polar/charged residues (Asn26, Glu27, Asp28, Arg34). The interaction with beta-arrestin exploits a greater circumference on the RAID1 helix, and involves two residues (Glu27, Asp28) that do not contribute to RACK1 binding. In contrast, the interaction of RACK1 with RAID1 is extended over a greater length of the helix and includes Leu37/Leu38, which do not contribute to beta-arrestin binding. A membrane-permeable, stearoylated Val12-Ser49 38-mer peptide disrupted the interaction of both beta-arrestin and RACK1 with endogenous PDE4D5 in HEKB2 cells, whilst a cognate peptide with a Glu27Ala substitution selectively failed to disrupt PDE4D5/RACK1 interaction. The stearoylated Val12-Ser49 38-mer peptide enhanced the isoprenaline-stimulated PKA phosphorylation of the beta(2)-adrenergic receptors (beta(2)AR) and its activation of ERK, whilst the Glu27Ala peptide was ineffective in both these regards.


Subject(s)
Arrestins/metabolism , GTP-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Phosphoric Diester Hydrolases/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Amino Acid Sequence , Arrestins/chemistry , Binding Sites , Cell Line , Circular Dichroism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/metabolism , GTP-Binding Proteins/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Isoproterenol/pharmacology , Models, Molecular , Molecular Sequence Data , Neoplasm Proteins/chemistry , Peptides/pharmacology , Phosphoric Diester Hydrolases/chemistry , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors for Activated C Kinase , Receptors, Adrenergic, beta-2/metabolism , Receptors, Cell Surface/chemistry , beta-Arrestins
9.
Biochem J ; 398(1): 23-36, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16689683

ABSTRACT

The cAMP-specific phosphodiesterase PDE4D5 can interact with the signalling scaffold proteins RACK (receptors for activated C-kinase) 1 and beta-arrestin. Two-hybrid and co-immunoprecipitation analyses showed that RACK1 and beta-arrestin interact with PDE4D5 in a mutually exclusive manner. Overlay studies with PDE4D5 scanning peptide array libraries showed that RACK1 and beta-arrestin interact at overlapping sites within the unique N-terminal region of PDE4D5 and at distinct sites within the conserved PDE4 catalytic domain. Screening scanning alanine substitution peptide arrays, coupled with mutagenesis and truncation studies, allowed definition of RACK1 and beta-arrestin interaction sites. Modelled on the PDE4D catalytic domain, these form distinct well-defined surface-exposed patches on helices-15-16, for RACK1, and helix-17 for beta-arrestin. siRNA (small interfering RNA)-mediated knockdown of RACK1 in HEK-293 (human embryonic kidney) B2 cells increased beta-arrestin-scaffolded PDE4D5 approx. 5-fold, increased PDE4D5 recruited to the beta2AR (beta2-adrenergic receptor) upon isoproterenol challenge approx. 4-fold and severely attenuated (approx. 4-5 fold) both isoproterenol-stimulated PKA (protein kinase A) phosphorylation of the beta2AR and activation of ERK (extracellular-signal-regulated kinase). The ability of a catalytically inactive form of PDE4D5 to exert a dominant negative effect in amplifying isoproterenol-stimulated ERK activation was ablated by a mutation that blocked the interaction of PDE4D5 with beta-arrestin. In the present study, we show that the signalling scaffold proteins RACK1 and beta-arrestin compete to sequester distinct 'pools' of PDE4D5. In this fashion, alterations in the level of RACK1 expression may act to modulate signal transduction mediated by the beta2AR.


Subject(s)
Arrestins/metabolism , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Peptide Library , Phosphoric Diester Hydrolases/metabolism , Protein Interaction Mapping , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Catalytic Domain/genetics , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Isoproterenol/pharmacology , Models, Molecular , Molecular Sequence Data , Phosphoric Diester Hydrolases/chemistry , Phosphorylation/drug effects , Protein Binding , RNA, Small Interfering/genetics , Receptors for Activated C Kinase , Receptors, Adrenergic, beta-2/metabolism , beta-Arrestins
10.
Cell Signal ; 17(9): 1158-73, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15905070

ABSTRACT

We employ a novel, dominant negative approach to identify a key role for certain tethered cyclic AMP specific phosphodiesterase-4 (PDE4) isoforms in regulating cyclic AMP dependent protein kinase A (PKA) sub-populations in resting COS1 cells. A fraction of PKA is clearly active in resting COS1 cells and this activity increases when cells are treated with the selective PDE4 inhibitor, rolipram. Point mutation of a critical, conserved aspartate residue in the catalytic site of long PDE4A4, PDE4B1, PDE4C2 and PDE4D3 isoforms renders them catalytically inactive. Overexpressed in resting COS1 cells, catalytically inactive forms of PDE4C2 and PDE4D3, but not PDE4A4 and PDE4B1, are constitutively PKA phosphorylated while overexpressed active versions of all these isoforms are not. Inactive and active versions of all these isoforms are PKA phosphorylated in cells where protein kinase A is maximally activated with forskolin and IBMX. By contrast, rolipram challenge of COS1 cells selectively triggers the PKA phosphorylation of recombinant, active PDE4D3 and PDE4C2 but not recombinant, active PDE4A4 and PDE4B1. Purified, recombinant PDE4D3 and PDE4A4 show a similar dose-dependency for in vitro phosphorylation by PKA. Disruption of the tethering of PKA type-II to PKA anchor proteins (AKAPs), achieved using the peptide Ht31, prevents inactive forms of PDE4C2 and PDE4D3 being constitutively PKA phosphorylated in resting cells as does siRNA-mediated knockdown of PKA-RII, but not PKA-RI. PDE4C2 and PDE4D3 co-immunoprecipitate from COS1 cell lysates with 250 kDa and 450 kDa AKAPs that tether PKA type-II and not PKA type-I. PKA type-II co-localises with AKAP450 in the centrosomal region of COS1 cells. The perinuclear distribution of recombinant, inactive PDE4D3, but not inactive PDE4A4, overlaps with AKAP450 and PKA type-II. The distribution of PKA phosphorylated inactive PDE4D3 also overlaps with that of AKAP450 in the centrosomal region of COS1 cells. We propose that a novel role for PDE4D3 and PDE4C2 is to gate the activation of AKAP450-tethered PKA type-II localised in the perinuclear region under conditions of basal cAMP generation in resting cells.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/physiology , Adaptor Proteins, Signal Transducing/metabolism , Centrosome/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/biosynthesis , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Activation , Isoenzymes/genetics , Isoenzymes/metabolism , Microscopy, Confocal , Mutation , Phosphodiesterase Inhibitors/pharmacology , RNA Interference , Rolipram/pharmacology
11.
J Biol Chem ; 278(49): 49230-8, 2003 Dec 05.
Article in English | MEDLINE | ID: mdl-14500724

ABSTRACT

Isoproterenol challenge of Hek-B2 cells causes a transient recruitment of the endogenous PDE4D isoforms found in these cells, namely PDE4D3 and PDE4D5, to the membrane fraction. PDE4D5 provides around 80% of the total PDE4D protein so recruited, although it only comprises about 40% of the total PDE4D protein in Hek-B2 cells. PDE4D5 provides about 80% of the total PDE4D protein found associated with beta-arrestins immunopurified from Hek-B2, COS1, and A549 cells as well as cardiac myocytes, whereas its overall level in these cells is between 15 and 50% of the total PDE4D protein. Truncation analyses indicate that two sites in PDE4D5 are involved in mediating its interaction with beta-arrestins, one associated with the common PDE4 catalytic region and the other located within its unique amino-terminal region. Truncation analyses indicate that two sites in beta-arrestin 2 are involved in mediating its interaction with PDE4D5, one associated with its extreme amino-terminal region and the other located within the carboxyl-terminal domain of the protein. We suggest that the unique amino-terminal region of PDE4D5 allows it to preferentially interact with beta-arrestins. This specificity appears likely to account for the preferential recruitment of PDE4D5, compared with PDE4D3, to membranes of Hek-B2 cells and cardiac myocytes upon challenge with isoproterenol.


Subject(s)
Arrestins/metabolism , Isoenzymes/metabolism , Phosphoric Diester Hydrolases/metabolism , Amino Acid Sequence , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Humans , Isoenzymes/chemistry , Molecular Sequence Data , Phosphoric Diester Hydrolases/chemistry , Protein Binding , beta-Arrestin 2 , beta-Arrestins
13.
BMC Biochem ; 3: 24, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12193273

ABSTRACT

BACKGROUND: The cyclic AMP specific phosphodiesterase, PDE4D5 interacts with the beta-propeller protein RACK1 to form a signaling scaffold complex in cells. Two-hybrid analysis of truncation and mutant constructs of the unique N-terminal region of the cAMP-specific phosphodiesterase, PDE4D5 were used to define a domain conferring interaction with the signaling scaffold protein, RACK1. RESULTS: Truncation and mutagenesis approaches showed that the RACK1-interacting domain on PDE4D5 comprised a cluster of residues provided by Asn-22/Pro-23/Trp-24/Asn-26 together with a series of hydrophobic amino acids, namely Leu-29, Val-30, Leu-33, Leu-37 and Leu-38 in a 'Leu-Xaa-Xaa-Xaa-Leu' repeat. This was done by 2-hybrid analyses and then confirmed in biochemical pull down analyses using GST-RACK1 and mutant PDE4D5 forms expressed in COS cells. Mutation of Arg-34, to alanine, in PDE4D5 attenuated its interaction with RACK1 both in 2-hybrid screens and in pull down analyses. A 38-mer peptide, whose sequence reflected residues 12 through 49 of PDE4D5, bound to RACK1 with similar affinity to native PDE4D5 itself (Ka circa 6 nM). CONCLUSIONS: The RACK1 Interaction Domain on PDE4D5, that we here call RAID1, is proposed to form an amphipathic helical structure that we suggest may interact with the C-terminal beta-propeller blades of RACK1 in a manner akin to the interaction of the helical G-gamma signal transducing protein with the beta-propeller protein, G-beta.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Cell Surface/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Binding Sites/physiology , COS Cells , CRADD Signaling Adaptor Protein , Chlorocebus aethiops , Cyclic AMP/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , GTP-Binding Proteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Phosphoric Diester Hydrolases/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors for Activated C Kinase , Receptors, Cell Surface/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...