Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Angiogenesis ; 16(1): 15-28, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22903372

ABSTRACT

The 5-hydroxytryptamine type 4 receptor (5-HT(4)R) regulates many physiological processes, including learning and memory, cognition, and gastrointestinal motility. Little is known about its role in angiogenesis. Using mouse hindlimb ischemia model of angiogenesis, we observed a significant reduction of limb blood flow recovery 14 days after ischemia and a decrease in density of CD31-positive vessels in adductor muscles in 5-HT(4)R(-/-) mice compared to wild type littermates. Our in vitro data indicated that 5-HT(4)R endogenously expressed in endothelial cells (ECs) may promote angiogenesis. Inhibition of the receptor with 5-HT(4)R antagonist RS 39604 reduced EC capillary tube formation in the reconstituted basement membrane. Using Boyden chamber migration assay and wound healing "scratch" assay, we demonstrated that RS 39604 treatment significantly suppressed EC migration. Transendothelial resistance measurement and immunofluorescence analysis showed that a 5-HT(4)R agonist RS 67333 led to an increase in endothelial permeability, actin stress fiber and interendothelial gap formation. Importantly, we provided the evidence that 5-HT(4)R-regulated EC migration may be mediated by Gα13 and RhoA. Our results suggest a prominent role of 5-HT(4)R in promoting angiogenesis and identify 5-HT(4)R as a potential therapeutic target for modulating angiogenesis under pathological conditions.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Receptors, Serotonin, 5-HT4/metabolism , Aniline Compounds/pharmacology , Animals , Capillaries/drug effects , Capillaries/growth & development , Cell Adhesion/drug effects , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Female , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Ischemia/metabolism , Ischemia/pathology , Mice , Muscles/blood supply , Muscles/pathology , Neovascularization, Physiologic/drug effects , Piperidines/pharmacology , Propane/analogs & derivatives , Propane/pharmacology , Regional Blood Flow/drug effects , Serotonin 5-HT4 Receptor Agonists/pharmacology , Serotonin 5-HT4 Receptor Antagonists/pharmacology , Wound Healing/drug effects , rhoA GTP-Binding Protein/metabolism
2.
Cell Signal ; 24(12): 2259-67, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22846544

ABSTRACT

Rhodopsin kinase (GRK1) is a member of G protein-coupled receptor kinase family and a key enzyme in the quenching of photolysed rhodopsin activity and desensitisation of the rod photoreceptor neurons. Like some other rod proteins involved in phototransduction, GRK1 is posttranslationally modified at the C terminus by isoprenylation (farnesylation), endoproteolysis and α-carboxymethylation. In this study, we examined the potential mechanisms of regulation of GRK1 methylation status, which have remained unexplored so far. We found that considerable fraction of GRK1 is endogenously methylated. In isolated rod outer segments, its methylation is inhibited and demethylation stimulated by low-affinity nucleotide binding. This effect is not specific for ATP and was observed in the presence of a non-hydrolysable ATP analogue AMP-PNP, GTP and other nucleotides, and thus may involve a site distinct from the active site of the kinase. GRK1 demethylation is inhibited in the presence of Ca(2+) by recoverin. This inhibition requires recoverin myristoylation and the presence of the membranes, and may be due to changes in GRK1 availability for processing enzymes upon its redistribution to the membranes induced by recoverin/Ca(2+). We hypothesise that increased GRK1 methylation in dark-adapted rods due to elevated cytoplasmic Ca(2+) levels would further increase its association with the membranes and recoverin, providing a positive feedback to efficiently suppress spurious phosphorylation of non-activated rhodopsin molecules and thus maximise senstivity of the photoreceptor. This study provides the first evidence for dynamic regulation of GRK1 α-carboxymethylation, which might play a role in the regulation of light sensitivity and adaptation in the rod photoreceptors.


Subject(s)
G-Protein-Coupled Receptor Kinase 1/metabolism , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Animals , Calcium/pharmacology , Cattle , Methylation/drug effects , Phosphorylation , Recoverin/pharmacology , Rod Cell Outer Segment/metabolism
3.
Funct Integr Genomics ; 12(1): 11-23, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21960277

ABSTRACT

PPP protein phosphatases are an important enzyme family involved in a variety of aspects of cellular signalling and metabolism. PPPs are ubiquitous in eukaryotes, and are also present in many bacteria. Canonical eukaryotic PPP phosphotases are represented by five major subfamilies (PP1, PP2A, calcineurin, PP5 and PPEF/PP7). We previously reported that three "bacterial-like" PPP groups span the prokaryote-eukaryote boundary, including "Shewanella-like" phosphatases (Shelphs), which are in the focus of this study. Here we predict possible biological functions and functional partners of Shelphs by examining composition of bacterial operons and expression data for eukaryotes available in public databases. In Arabidopsis thaliana, the predicted possible roles include light-dependent regulation of chloroplast functions, signalling between the nucleus and the chloroplast, and defence responses. In Plasmodium falciparum, Shelphs are predicted to be associated with host cell invasion. One isoform has been located in the apical complex, essential for the interaction with the host cell. This makes P. falciparum Shelphs obvious potential candidates for therapeutic targets. Shelphs are also present in bacteria that constitute a considerable proportion of symbiotic microflora in humans. The predicted involvement of bacterial Shelphs in sensing and import of nutrients and extrusion of toxins may be relevant to the links between physiology of humans and our symbionts. Thus, despite the absence of Shelphs in animals, including humans, they may have a direct relationship to human health. Some predicted biological processes and potential functional partners of Shelphs are common between different bacterial and/or eukaryotic lineages, suggesting evolutionary conservation of some Shelph regulatory modules.


Subject(s)
Bacterial Proteins/physiology , Phosphoprotein Phosphatases/physiology , Shewanella/enzymology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/physiology , Gene Expression Regulation, Enzymologic , Humans , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Proteins/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/physiology
4.
J Cell Physiol ; 226(5): 1255-64, 2011 May.
Article in English | MEDLINE | ID: mdl-20945373

ABSTRACT

Vasodilator-stimulated phosphoprotein (VASP) is implicated in the protection of the endothelial barrier in vitro and in vivo. The function of VASP in thrombin signaling in the endothelial cells (ECs) is not known. For the first time we studied the effects of VASP deficiency on EC permeability and pulmonary vascular permeability in response to thrombin receptor stimulation. We provided the evidence that VASP deficiency potentiates the increase in endothelial permeability induced by activation of thrombin receptor in cultured human umbilical vein endothelial cells (HUVECs) and isolated mouse lungs. Using transendothelial resistance measurement, we showed that siRNA-mediated VASP downregulation in HUVECs leads to a potentiation of thrombin- and protease-activated receptor 1 (PAR-1) agonist-induced increase in endothelial permeability. Compared to control cells, VASP-deficient HUVECs had delayed endothelial junctional reassembly and abrogated VE-cadherin cytoskeletal anchoring in the recovery phase after thrombin stimulation, as demonstrated by immunofluorescence studies and cell fractionation analysis, respectively. Measurement of the capillary filtration coefficient in isolated mouse lungs demonstrated that VASP(-/-) mice have increased microvascular permeability in response to infusion with PAR-1 agonist compared to wild type mice. Lack of VASP led to decreased Rac1 activation both in VASP-deficient HUVECs after thrombin stimulation and VASP(-/-) mouse lungs after PAR-1 agonist infusion, indicating that VASP effects on thrombin signaling may be correlated with changes in Rac1 activity. This study demonstrates that VASP may play critical and complex role in the regulation of thrombin-dependent disruption of the endothelial barrier function.


Subject(s)
Capillary Permeability , Cell Adhesion Molecules/deficiency , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Lung/blood supply , Microfilament Proteins/deficiency , Phosphoproteins/deficiency , Receptor, PAR-1/metabolism , Thrombin/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Adhesion Molecules/genetics , Cells, Cultured , Electric Impedance , Humans , Intercellular Junctions/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Neuropeptides/metabolism , Phosphoproteins/genetics , RNA Interference , Time Factors , Transfection , Up-Regulation , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism
5.
Int J Biochem Cell Biol ; 42(11): 1816-22, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20674765

ABSTRACT

The function of protein phosphatases with EF-hand domains (PPEF) in mammals is not known. Large-scale expression profiling experiments suggest that PPEF expression may correlate with stress protective responses, cell survival, growth, proliferation, or neoplastic transformation. Apoptosis signal regulating kinase-1 (ASK1) is a MAP kinase kinase kinase implicated in cancer, cardiovascular and neurodegenerative diseases. ASK1 is activated by oxidative stress and induces pro-apoptotic or inflammatory signalling, largely via sustained activation of MAP kinases p38 and/or JNK. We identify human PPEF2 as a novel interacting partner and a negative regulator of ASK1. In COS-7 or HEK 293A cells treated with H(2)O(2), expression of PPEF2 abrogated sustained activation of p38 and one of the JNK p46 isoforms, and prevented ASK1-dependent caspase-3 cleavage and activation. PPEF2 efficiently suppressed H(2)O(2)-induced activation of ASK1. Overexpessed as well as endogenous ASK1 co-immunoprecipitated with PPEF2. PPEF2 was considerably more potent both as a suppressor of ASK1 activation and as its interacting partner as compared to protein phosphatase 5 (PP5), a well-known negative regulator of ASK1. PPEF2 was found to form complexes with endogenous Hsp70 and to a lesser extent Hsp90, which are also known interacting partners of PP5. These data identify, for the first time, a possible downstream signalling partner of a mammalian PPEF phosphatase, and suggest that, despite structural divergence, PPEF and PP5 phosphatases may share common interacting partners and functions.


Subject(s)
MAP Kinase Kinase Kinase 5/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , COS Cells , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Chlorocebus aethiops , EF Hand Motifs/genetics , EF Hand Motifs/physiology , Humans , Hydrogen Peroxide/pharmacology , Immunoblotting , Immunoprecipitation , MAP Kinase Kinase Kinase 5/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Phosphoprotein Phosphatases/genetics , Protein Binding
6.
Genes Chromosomes Cancer ; 49(9): 775-90, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20607704

ABSTRACT

We review the evidence suggesting the involvement of Cadherin 13 (CDH13, T-cadherin, H-cadherin) in various cancers. CDH13 is an atypical member of the cadherin family, devoid of a transmembrane domain and anchored to the exterior surface of the plasma membrane via a glycosylphosphatidylinositol anchor. CDH13 is thought to affect cellular behavior largely through its signaling properties. It is often down-regulated in cancerous cells. CDH13 down-regulation has been associated with poorer prognosis in various carcinomas, such as lung, ovarian, cervical and prostate cancer. CDH13 re-expression in most cancer cell lines inhibits cell proliferation and invasiveness, increases susceptibility to apoptosis, and reduces tumor growth in in vivo models. These properties suggest that CDH13 may represent a possible target for therapy in some cancers. At the same time, CDH13 is up-regulated in blood vessels growing through tumors and promotes tumor neovascularization. In contrast to most cancer cell lines, CDH13 overexpression in endothelial cells promotes their proliferation and migration, and has a pro-survival effect. We also discuss molecular mechanisms that may regulate CDH13 expression and underlie its roles in cancer.


Subject(s)
Cadherins/physiology , Genes, Tumor Suppressor , Neoplasms/pathology , Cell Movement , Cell Proliferation , Humans
7.
J Cell Physiol ; 223(1): 94-102, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20039275

ABSTRACT

T-cadherin is an atypical member of the cadherin family, which lacks the transmembrane and intracellular domains and is attached to the plasma membrane via a glycosylphosphatidylinositol anchor. Unlike canonical cadherins, it is believed to function primarily as a signaling molecule. T-cadherin is highly expressed in endothelium. Using transendothelial electrical resistance measurements and siRNA-mediated depletion of T-cadherin in human umbilical vein endothelial cells, we examined its involvement in regulation of endothelial barrier. We found that in resting confluent monolayers adjusted either to 1% or 10% serum, T-cadherin depletion modestly, but consistently reduced transendothelial resistance. This was accompanied by increased phosphorylation of Akt and LIM kinase, reduced phosphorylation of p38 MAP kinase, but no difference in tubulin acetylation and in phosphorylation of an actin filament severing protein cofilin and myosin light chain kinase. Serum stimulation elicited a biphasic increase in resistance with peaks at 0.5 and 4-5 h, which was suppressed by a PI3 kinase/Akt inhibitor wortmannin and a p38 inhibitor SB 239063. T-cadherin depletion increased transendothelial resistance between the two peaks and reduced the amplitude of the second peak. T-cadherin depletion abrogated serum-induced Akt phosphorylation at Thr308 and reduced phosphorylation at Ser473, reduced phosphorylation of cofilin, and accelerated tubulin deacetylation. Adiponectin slightly improved transendothelial resistance irrespectively of T-cadherin depletion. T-cadherin depletion also resulted in a reduced sensitivity and delayed responses to thrombin. These data implicate T-cadherin in regulation of endothelial barrier function, and suggest a complex signaling network that links T-cadherin and regulation of barrier function.


Subject(s)
Cadherins/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Acetylation , Actin Depolymerizing Factors/metabolism , Adiponectin/metabolism , Cadherins/genetics , Capillary Permeability/drug effects , Cardiac Myosins/metabolism , Cells, Cultured , Electric Impedance , Endothelial Cells/drug effects , Humans , Lim Kinases/metabolism , Myosin Light Chains/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction , Thrombin/metabolism , Time Factors , Tubulin/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Am J Physiol Cell Physiol ; 297(5): C1168-77, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726744

ABSTRACT

T-cadherin (H-cadherin, cadherin 13) is upregulated in vascular proliferative disorders and in tumor-associated neovascularization and is deregulated in many cancers. Unlike canonical cadherins, it lacks transmembrane and intracellular domains and is attached to the plasma membrane via a glycosylphosphatidylinositol anchor. T-cadherin is thought to function in signaling rather than as an adhesion molecule. Some interactive partners of T-cadherin at the plasma membrane have recently been identified. We examined T-cadherin location in human endothelial cells using confocal microscopy and subcellular fractionation. We found that a considerable proportion of T-cadherin is located in the nucleus and in the centrosomes. T-cadherin colocalized with a centrosomal marker gamma-tubulin uniformly throughout the cell cycle at least in human umbilical vein endothelial cells. In the telophase, T-cadherin transiently concentrated in the midbody and was apparently degraded. Its overexpression resulted in an increase in the number of multinuclear cells, whereas its downregulation by small interfering RNA led to an increase in the number of cells with multiple centrosomes. These findings indicate that deregulation of T-cadherin in endothelial cells may lead to disturbances in cytokinesis or centrosomal replication.


Subject(s)
Cadherins/metabolism , Cell Nucleus/metabolism , Centrosome/metabolism , Cytokinesis/physiology , Endothelial Cells/metabolism , Blotting, Western , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Microscopy, Confocal , Protein Transport/physiology , RNA, Small Interfering , Tubulin/metabolism
9.
Cell Mol Life Sci ; 66(19): 3103-10, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19662497

ABSTRACT

PPEF/PP7 represents one of the five subfamilies of the PPP protein Ser/Thr phosphatases. Studies published in recent years point to a role of plant PP7 at a crossroad of different pathways of light and stress signalling. In animals, PPEFs are highly expressed in sensory neurons, and Drosophila PPEF phosphatase, rdgC, is essential for dephosphorylation of rhodopsin. Expression profiling suggests that mammalian PPEF may play a role in stress-protective responses, cell survival, growth, proliferation, and oncogenesis. Despite structural similarities of the catalytic domains and the fact that some of these phosphatases are involved in light perception both in animals and in plants, the plant and non-plant representatives of this group have distinct domain architecture and appear not to be orthologues.


Subject(s)
Phosphoprotein Phosphatases/physiology , Animals , Binding Sites , EF Hand Motifs , Humans , Phosphoprotein Phosphatases/chemistry , Plant Proteins/chemistry , Plant Proteins/physiology , Protein Structure, Tertiary
10.
Mol Biochem Parasitol ; 161(2): 81-90, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18619495

ABSTRACT

Protein phosphorylation is an important mechanism implicated in physiology of any organism, including parasitic protozoa. Enzymes that control protein phosphorylation (kinases and phosphatases) are considered promising targets for drug development. This review attempts to provide the first account of the current understanding of the structure, regulation and biological functions of protein Ser/Thr phosphatases in unicellular parasites. We have examined the complements of phosphatases ("phosphatomes") of the PPP and PPM families in several species of Apicomplexa (including malaria parasite Plasmodium), as well as Giardia lamblia, Entamoeba histolytica, Trichomonas vaginalis and a microsporidium Encephalitozoon cuniculi. Apicomplexans have homologues (in most cases represented by single isoforms) of all human PPP subfamilies. Some apicomplexan PPP phosphatases have no orthologues in their vertebrate hosts, including a previously unrecognised group of pseudo-phosphatases with putative Ca(2+)-binding domains, which we designate as EFPP. We also describe the presence of previously undetected Zn finger motifs in PPEF phosphatases from kinetoplastids, and a likely case of convergent evolution of tetratricopeptide repeat domain-containing phosphatases in G. lamblia. Among the parasites examined, E. cuniculi has the smallest Ser/Thr phosphatome (5 PPP and no PPM), while T. vaginalis shows the largest expansion of the PPP family (169 predicted phosphatases). Most protozoan PPM phosphatases cluster separately from human sequences. The structural peculiarities or absence of human orthologues of a number of protozoan protein Ser/Thr phosphatases makes them potentially suitable targets for chemotherapy and thus warrants their functional assessment.


Subject(s)
Eukaryota/enzymology , Phosphoprotein Phosphatases , Animals , Eukaryota/classification , Eukaryota/genetics , Evolution, Molecular , Humans , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phylogeny , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
11.
Int J Parasitol ; 38(11): 1279-95, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18547579

ABSTRACT

The aim of this review is to provide a synthesis of the published experimental data on protein tyrosine phosphatases from parasitic protozoa, in silico analysis based on the availability of completed genomes and to place available data for individual phosphatases from different unicellular parasites into the comparative and evolutionary context. We analysed the complement of protein tyrosine phosphatases (PTP) in several species of unicellular parasites that belong to Apicomplexa (Plasmodium; Cryptosporidium, Babesia, Theileria, and Toxoplasma), kinetoplastids (Leishmania and Trypanosoma spp.), as well as Entamoeba histolytica, Giardia lamblia, Trichomonas vaginalis and a microsporidium Encephalitozoon cuniculi. The analysis shows distinct distribution of the known families of tyrosine phosphatases in different species. Protozoan tyrosine phosphatases show considerable levels of divergence compared with their mammalian homologues, both in terms of sequence similarity between the catalytic domains and the structure of their flanking domains. This potentially makes them suitable targets for development of specific inhibitors with minimal effects on physiology of mammalian hosts.


Subject(s)
Eukaryota/enzymology , Protein Tyrosine Phosphatases , Animals , Apicomplexa/enzymology , Kinetoplastida/enzymology , Microsporidia, Unclassified/immunology , Phosphorylation , Protein Tyrosine Phosphatases/classification , Protein Tyrosine Phosphatases/metabolism , RNA, Protozoan/metabolism
12.
FASEB J ; 22(8): 2821-31, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18367648

ABSTRACT

G alpha12 constitutes, along with G alpha13, one of the four families of alpha subunits of heterotrimeric G proteins. We found that the N terminus of G alpha12, but not those of other G alpha subunits, contains a predicted mitochondrial targeting sequence. Using confocal microscopy and cell fractionation, we demonstrated that up to 40% of endogenous G alpha12 in human umbilical vein endothelial cells colocalize with mitochondrial markers. N-terminal sequence of G alpha12 fused to GFP efficiently targeted the fusion protein to mitochondria. G alpha12 with mutated mitochondrial targeting sequence was still located in mitochondria, suggesting the existence of additional mechanisms for mitochondrial localization. Lysophosphatidic acid, one of the known stimuli transduced by G alpha12/13, inhibited mitochondrial motility, while depletion of endogenous G alpha12 increased mitochondrial motility. G alpha12Q229L variants uncoupled from RhoGEFs (but not fully functional activated G alpha12Q229L) induced transformation of the mitochondrial network into punctate mitochondria and resulted in a loss of mitochondrial membrane potential. All examined G alpha12Q229L variants reduced phosphorylation of Bcl-2 at Ser-70, while only mutants unable to bind RhoGEFs also decreased cellular levels of Bcl-2. These G alpha12 mutants were also more efficient Hsp90 interactors. These findings are the first demonstration of a heterotrimeric G protein alpha subunit specifically targeted to mitochondria and involved in the control of mitochondrial morphology and dynamics.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Mitochondria/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Guanine Nucleotide Exchange Factors/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Potential, Mitochondrial , Movement , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors
13.
J Mol Signal ; 2: 13, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17971232

ABSTRACT

Heterotrimeric G proteins are ubiquitous signaling partners of seven transmembrane-domain G-protein-coupled receptors (GPCRs), the largest (and most important pharmacologically) receptor family in mammals. A number of scaffolding proteins have been identified that regulate various facets of GPCR signaling. In this review, we summarize current knowledge concerning those scaffolding proteins that are known to directly bind heterotrimeric G proteins, and discuss the composition of the protein complexes they assemble and their effects on signal transduction. Emerging evidence about possible ways of regulation of activity of these scaffolding proteins is also discussed.

14.
FASEB J ; 21(13): 3727-36, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17595347

ABSTRACT

Apoptosis signal-regulating kinase (ASK1) is a mitogen-activated protein kinase (MAPK) that transduces apoptotic signals from a variety of stresses. We have shown previously that alpha subunits of heterotrimeric G12 and G13 proteins stimulate ASK1 kinase activity and ASK1-dependent apoptosis. Here, we report a novel mechanism of G-protein-dependent regulation of ASK1. We demonstrated that G alpha13 forms a complex with ASK1 in an activation-independent manner. Both N- and C-terminal regulatory domains of ASK1 were essential for the efficient interaction, while its kinase domain was not required. Formation of the G alpha13-ASK1 complex was enhanced by JNK-interacting leucine zipper protein, JLP. Constitutively activated G alpha13Q226L increased ASK1 expression. Short-term activation of a serotonin 5-HT4 receptor that is coupled to G alpha13 also increased ASK1 expression. Importantly, prolonged activation of 5-HT4 receptor in COS-7 cells or prolonged treatment of human umbilical vein endothelial cells with thrombin concomitantly down-regulated both G alpha13 and ASK1. Data showed that G alpha13Q226L reduced the rate of ASK1 degradation, decreased ASK1 ubiquitination, and reduced association of ASK1 with an E3 ubiquitin ligase CHIP, previously shown to mediate ASK1 degradation. Our findings indicate that ASK1 expression levels can be regulated by G alpha13, at least in part via control of ASK1 ubiquitination and degradation.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/physiology , MAP Kinase Kinase Kinase 5/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Hydrolysis
15.
Am J Physiol Lung Cell Mol Physiol ; 293(2): L259-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17496061

ABSTRACT

Molecular mechanisms of surfactant delivery to the air/liquid interface in the lung, which is crucial to lower the surface tension, have been studied for more than two decades. Lung surfactant is synthesized in the alveolar type II cells. Its delivery to the cell surface is preceded by surfactant component synthesis, packaging into specialized organelles termed lamellar bodies, delivery to the apical plasma membrane and fusion. Secreted surfactant undergoes reuptake, intracellular processing, and finally resecretion of recycled material. This review focuses on the mechanisms of delivery of surfactant components to and their secretion from lamellar bodies. Lamellar bodies-independent secretion is also considered. Signal transduction pathways involved in regulation of these processes are discussed as well as disorders associated with their malfunction.


Subject(s)
Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Pulmonary Surfactants/metabolism , Signal Transduction/physiology , Animals , Humans
16.
Expert Opin Ther Targets ; 10(5): 723-33, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16981829

ABSTRACT

Alpha soluble NSF attachment protein (alphaSNAP) is a ubiquitous and indispensable component of membrane fusion machinery. Deletion of alphaSNAP is embryonically lethal. Yet, there is accumulating evidence that milder alterations in expression levels of alphaSNAP may be associated with a number of specific pathological conditions, such as several neurological disorders, Type 2 diabetes and aggressive neuroendocrine tumours. Here, the authors review the evidence available for animal models and for humans, and discuss possible therapeutic approaches that may target alphaSNAP.


Subject(s)
Diabetes Mellitus/metabolism , Drug Delivery Systems/methods , Membrane Fusion Proteins/metabolism , Neoplasms/metabolism , Nervous System Diseases/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Animals , Diabetes Mellitus/drug therapy , Humans , Neoplasms/drug therapy , Nervous System Diseases/drug therapy , Protein Isoforms/metabolism , SNARE Proteins/administration & dosage , SNARE Proteins/metabolism
17.
Mol Pharmacol ; 69(3): 975-82, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16326932

ABSTRACT

Endothelial nitric-oxide synthase (eNOS) plays a crucial role in the regulation of a variety of cardiovascular and pulmonary functions in both normal and pathological conditions. Multiple signaling inputs, including calcium, caveolin-1, phosphorylation by several kinases, and binding to the 90-kDa heat shock protein (Hsp90), regulate eNOS activity. Here, we report a novel mechanism of G protein-dependent regulation of eNOS. We demonstrate that in mammalian cells, the alpha subunit of heterotrimeric G12 protein (G alpha12) can form a complex with eNOS in an activation- and Hsp90-independent manner. Our data show that G alpha12 does not affect eNOS-specific activity, but it strongly enhances total eNOS activity by increasing cellular levels of eNOS. Experiments using inhibition of protein or mRNA synthesis show that G alpha12 increases the expression of eNOS by increasing half-life of both eNOS protein and eNOS mRNA. Small interfering RNA-mediated depletion of endogenous G alpha12 decreases eNOS levels. A quantitative correlation can be detected between the extent of down-regulation of G alpha12 and eNOS in endothelial cells after prolonged treatment with thrombin. G protein-dependent increase of eNOS expression represents a novel mechanism by which heterotrimeric G proteins can regulate the activity of downstream signaling molecules.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Activation , Enzyme Stability , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, G12-G13/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Nitric Oxide Synthase Type III/genetics , RNA Stability , RNA, Messenger/analysis , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Thrombin/pharmacology
18.
J Biol Chem ; 280(34): 30376-83, 2005 Aug 26.
Article in English | MEDLINE | ID: mdl-15980433

ABSTRACT

The involvement of heterotrimeric G proteins in the regulation of adherens junction function is unclear. We identified alphaSNAP as an interactive partner of G alpha12 using yeast two-hybrid screening. Glutathione S-transferase pull-down assays showed the selective interaction of alphaSNAP with G alpha12 in COS-7 as well as in human umbilical vein endothelial cells. Using domain swapping experiments, we demonstrated that the N-terminal region of G alpha12 (1-37 amino acids) was necessary and sufficient for its interaction with alphaSNAP. G alpha13 with its N-terminal extension replaced by that of G alpha12 acquired the ability to bind to alphaSNAP, whereas G alpha12 with its N terminus replaced by that of G alpha13 lost this ability. Using four point mutants of alphaSNAP, which alter its ability to bind to the SNARE complex, we determined that the convex rather than the concave surface of alphaSNAP was involved in its interaction with G alpha12. Co-transfection of human umbilical vein endothelial cells with G alpha12 and alphaSNAP stabilized VE-cadherin at the plasma membrane, whereas down-regulation of alphaSNAP with siRNA resulted in the loss of VE-cadherin from the cell surface and, when used in conjunction with G alpha12 overexpression, decreased endothelial barrier function. Our results demonstrate a direct link between the alpha subunit of G12 and alphaSNAP, an essential component of the membrane fusion machinery, and implicate a role for this interaction in regulating the membrane localization of VE-cadherin and endothelial barrier function.


Subject(s)
Cadherins/metabolism , Endothelium, Vascular/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Vesicular Transport Proteins/chemistry , Amino Acid Sequence , Animals , Antigens, CD , COS Cells , Cell Membrane/metabolism , Cells, Cultured , GTP-Binding Protein alpha Subunits, G12-G13/chemistry , Gene Expression Regulation , Glutathione Transferase/metabolism , Glycerol/pharmacology , Humans , Immunoblotting , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins , Transfection , Two-Hybrid System Techniques , Umbilical Veins/cytology , Vesicular Transport Proteins/metabolism
19.
J Biol Chem ; 280(27): 25388-95, 2005 Jul 08.
Article in English | MEDLINE | ID: mdl-15890660

ABSTRACT

Recent evidence has implicated the protein phosphatase PP5 in a variety of signaling pathways. Whereas several proteins have been identified that interact with PP5 and regulate its activity, a possibility of its regulation by second messengers remains speculative. Activation of PP5 in vitro by polyunsaturated fatty acids (e.g. arachidonic acid) and fatty acyl-CoA esters (e.g. arachidonoyl-CoA) has been reported. We report here that PP5 is strongly inhibited by micromolar concentrations of a natural polyamine spermine. This inhibition was observed both in assays with a low molecular weight substrate p-nitrophenyl phosphate as well as phosphocasein and apoptosis signal-regulating kinase 1 (ASK1), thought to be a physiological substrate of PP5. Furthermore, a decrease in polyamine levels in COS-7 cells induced by alpha-difluoromethylornithine (DFMO), an inhibitor of ornithine decarboxylase, led to accelerated dephosphorylation of oxidative stress-activated ASK1. This effect was suppressed by okadaic acid and by siRNA-mediated PP5 depletion, indicating that the effect of polyamine levels on ASK1 dephosphorylation was mediated by PP5. In line with the decreased ASK1 activation, polyamine depletion in COS-7 cells abrogated oxidative stress-induced activation of caspase-3, which executes ASK1-induced apoptosis, as well as caspase-3 activation induced by ASK1 overexpression, but had no effect on basal caspase-3 activity. These results implicate polyamines, emerging intracellular signaling molecules, as potential physiological regulators of PP5. Our findings also suggest a novel mechanism of the anti-apoptotic action of a decrease in polyamine levels via de-inhibition of PP5 and accelerated dephosphorylation and deactivation of ASK1.


Subject(s)
Apoptosis/physiology , MAP Kinase Kinase Kinase 5/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Spermine/metabolism , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation/physiology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Oxidative Stress/physiology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Phosphorylation , RNA, Small Interfering , Signal Transduction/physiology , Substrate Specificity
20.
BMC Evol Biol ; 4: 47, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15555063

ABSTRACT

BACKGROUND: In eukaryotes, PPP (protein phosphatase P) family is one of the two known protein phosphatase families specific for Ser and Thr. The role of PPP phosphatases in multiple signaling pathways in eukaryotic cell has been extensively studied. Unlike eukaryotic PPP phosphatases, bacterial members of the family have broad substrate specificity or may even be Tyr-specific. Moreover, one group of bacterial PPPs are diadenosine tetraphosphatases, indicating that bacterial PPP phosphatases may not necessarily function as protein phosphatases. RESULTS: We describe the presence in eukaryotes of three groups of expressed genes encoding "non-conventional" phosphatases of the PPP family. These enzymes are more closely related to bacterial PPP phosphatases than to the known eukaryotic members of the family. One group, found exclusively in land plants, is most closely related to PPP phosphatases from some alpha-Proteobacteria, including Rhizobiales, Rhodobacterales and Rhodospirillaceae. This group is therefore termed Rhizobiales / Rhodobacterales / Rhodospirillaceae-like phosphatases, or Rhilphs. Phosphatases of the other group are found in Viridiplantae, Rhodophyta, Trypanosomatidae, Plasmodium and some fungi. They are structurally related to phosphatases from psychrophilic bacteria Shewanella and Colwellia, and are termed Shewanella-like phosphatases, or Shelphs. Phosphatases of the third group are distantly related to ApaH, bacterial diadenosine tetraphosphatases, and are termed ApaH-like phosphatases, or Alphs. Patchy distribution of Alphs in animals, plants, fungi, diatoms and kinetoplasts suggests that these phosphatases were present in the common ancestor of eukaryotes but were independently lost in many lineages. Rhilphs, Shelphs and Alphs form PPP clades, as divergent from "conventional" eukaryotic PPP phosphatases as they are from each other and from major bacterial clades. In addition, comparison of primary structures revealed a previously unrecognised (I/L/V)D(S/T)G motif, conserved in all bacterial and "bacterial-like" eukaryotic PPPs, but not in "conventional" eukaryotic and archaeal PPPs. CONCLUSIONS: Our findings demonstrate that many eukaryotes possess diverse "bacterial-like" PPP phosphatases, the enzymatic characteristics, physiological roles and precise evolutionary history of which have yet to be determined.


Subject(s)
Bacterial Proteins/chemistry , Eukaryotic Cells/enzymology , Phosphoprotein Phosphatases/chemistry , Alphaproteobacteria/enzymology , Amino Acid Motifs/genetics , Amino Acid Sequence/genetics , Animals , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Bacterial Proteins/genetics , Chlamydomonas reinhardtii/enzymology , Molecular Sequence Data , Phosphoprotein Phosphatases/genetics , Protein Structure, Tertiary/genetics , Schizosaccharomyces pombe Proteins/genetics , Shewanella/enzymology , Ticks/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...