Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(19)2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36232837

ABSTRACT

CCG-1423 is a Rho A pathway inhibitor that has been reported to inhibit Rho/SRF-mediated transcriptional regulation. Serum response factor and its cofactors, which include ternary complex factors and myocardin-related transcription factors, regulate various cellular functions. In this study, we observed that CCG-1423 modulates the mitochondrial functions. The effect of this small molecule drug was determined by measuring mitochondrial function using an XFe96 Analyzer and an Oxygraph 2k (O2k) high-resolution respirometer. CCG-1423 treatment significantly reduced oxidative phosphorylation in a dose-dependent manner. However, CCG-1423 increased the glycolytic rate. We also observed that histone 4 at lysine-16 underwent hyperacetylation with the treatment of this drug. Immunolabeling with F-actin and MitoTracker revealed the alteration in the actin cytoskeleton and mitochondria. Taken together, our findings highlight a critical role of CCG-1423 in inhibiting the transcription of SRF/p49 and PGC-1α, ß, resulting in the downregulation of mitochondrial genes, leading to the repression of mitochondrial oxidative phosphorylation and overall ATP reduction. This study provides a better understanding of the effects of CCG-1423 on mitochondria, which may be useful for the assessment of the potential clinical application of CCG-1423 and its derivatives.


Subject(s)
Actins , Serum Response Factor , Actins/metabolism , Adenosine Triphosphate , Anilides , Benzamides , Histones , Lysine , Mitochondria/metabolism , Ternary Complex Factors/metabolism , Transcription Factors/metabolism
2.
Elife ; 112022 01 19.
Article in English | MEDLINE | ID: mdl-35044299

ABSTRACT

Serum response factor (SRF) is an essential transcription factor that influences many cellular processes including cell proliferation, migration, and differentiation. SRF directly regulates and is required for immediate early gene (IEG) and actin cytoskeleton-related gene expression. SRF coordinates these competing transcription programs through discrete sets of cofactors, the ternary complex factors (TCFs) and myocardin-related transcription factors (MRTFs). The relative contribution of these two programs to in vivo SRF activity and mutant phenotypes is not fully understood. To study how SRF utilizes its cofactors during development, we generated a knock-in SrfaI allele in mice harboring point mutations that disrupt SRF-MRTF-DNA complex formation but leave SRF-TCF activity unaffected. Homozygous SrfaI/aI mutants die at E10.5 with notable cardiovascular phenotypes, and neural crest conditional mutants succumb at birth to defects of the cardiac outflow tract but display none of the craniofacial phenotypes associated with complete loss of SRF in that lineage. Our studies further support an important role for MRTF mediating SRF function in cardiac neural crest and suggest new mechanisms by which SRF regulates transcription during development.


Subject(s)
Neural Crest/embryology , Serum Response Factor/genetics , Ternary Complex Factors/genetics , Transcription Factors/genetics , Animals , Mice , Serum Response Factor/metabolism , Ternary Complex Factors/metabolism , Transcription Factors/metabolism
3.
Nat Commun ; 12(1): 6829, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819513

ABSTRACT

Signals are relayed from receptor tyrosine kinases (RTKs) at the cell surface to effector systems in the cytoplasm and nucleus, and coordination of this process is important for the execution of migratory phenotypes, such as cell scattering and invasion. The endosomal system influences how RTK signalling is coded, but the ways in which it transmits these signals to the nucleus to influence gene expression are not yet clear. Here we show that hepatocyte growth factor, an activator of MET (an RTK), promotes Rab17- and clathrin-dependent endocytosis of EphA2, another RTK, followed by centripetal transport of EphA2-positive endosomes. EphA2 then mediates physical capture of endosomes on the outer surface of the nucleus; a process involving interaction between the nuclear import machinery and a nuclear localisation sequence in EphA2's cytodomain. Nuclear capture of EphA2 promotes RhoG-dependent phosphorylation of the actin-binding protein, cofilin to oppose nuclear import of G-actin. The resulting depletion of nuclear G-actin drives transcription of Myocardin-related transcription factor (MRTF)/serum-response factor (SRF)-target genes to implement cell scattering and the invasive behaviour of cancer cells.


Subject(s)
Cell Nucleus/metabolism , Endosomes/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/pathology , Ternary Complex Factors/metabolism , Actins/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Cell Line, Tumor , Cytoplasm/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Mice , Mice, Knockout , Neoplasm Invasiveness/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism
4.
FEBS J ; 288(10): 3120-3134, 2021 05.
Article in English | MEDLINE | ID: mdl-32885587

ABSTRACT

Serum response factor (SRF), a member of the Mcm1, Agamous, Deficiens, and SRF (MADS) box transcription factor, is widely expressed in all cell types and plays a crucial role in the physiological function and development of diseases. SRF regulates its downstream genes by binding to their CArG DNA box by interacting with various cofactors. However, the underlying mechanisms are not fully understood, therefore attracting increasing research attention due to the importance of this topic. This review's objective is to discuss the new progress in the studies of the molecular mechanisms involved in the activation of SRF and its impacts in physiological and pathological conditions. Notably, we summarized the recent studies on the interaction of SRF with its two main types of cofactors belonging to the myocardin families of transcription factors and the members of the ternary complex factors. The knowledge of these mechanisms will create new opportunities for understanding the dynamics of many traits and disease pathogenesis especially, cardiovascular diseases and cancer that could serve as targets for pharmacological control and treatment of these diseases.


Subject(s)
Cardiovascular Diseases/genetics , Neoplasms/genetics , Nuclear Proteins/genetics , Serum Response Factor/genetics , Ternary Complex Factors/genetics , Trans-Activators/genetics , Transcription, Genetic , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Apoptosis/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cell Proliferation , DNA/genetics , DNA/metabolism , Gene Expression Regulation , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Serum Response Factor/metabolism , Signal Transduction , Stress, Physiological , Ternary Complex Factors/metabolism , Trans-Activators/metabolism
5.
Science ; 368(6489)2020 04 24.
Article in English | MEDLINE | ID: mdl-32327570

ABSTRACT

Protein quality control is essential for the proper function of cells and the organisms that they make up. The resulting loss of proteostasis, the processes by which the health of the cell's proteins is monitored and maintained at homeostasis, is associated with a wide range of age-related human diseases. Here, we highlight how the integrated stress response (ISR), a central signaling network that responds to proteostasis defects by tuning protein synthesis rates, impedes the formation of long-term memory. In addition, we address how dysregulated ISR signaling contributes to the pathogenesis of complex diseases, including cognitive disorders, neurodegeneration, cancer, diabetes, and metabolic disorders. The development of tools through which the ISR can be modulated promises to uncover new avenues to diminish pathologies resulting from it for clinical benefit.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Proteostasis , Stress, Physiological , Ternary Complex Factors/metabolism , Acetamides/chemistry , Acetamides/pharmacology , Animals , Cyclohexylamines/chemistry , Cyclohexylamines/pharmacology , Eukaryotic Initiation Factor-2/antagonists & inhibitors , Humans , Immunity , Metabolic Diseases/metabolism , Mice , Neoplasms/metabolism , Phosphotransferases/metabolism
6.
Proc Natl Acad Sci U S A ; 115(11): E2653-E2662, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29487210

ABSTRACT

There is currently an unmet need for versatile techniques to monitor the assembly and dynamics of ternary complexes in live cells. Here we describe bioluminescence resonance energy transfer with fluorescence enhancement by combined transfer (BRETFect), a high-throughput technique that enables robust spectrometric detection of ternary protein complexes based on increased energy transfer from a luciferase to a fluorescent acceptor in the presence of a fluorescent intermediate. Its unique donor-intermediate-acceptor relay system is designed so that the acceptor can receive energy either directly from the donor or indirectly via the intermediate in a combined transfer, taking advantage of the entire luciferase emission spectrum. BRETFect was used to study the ligand-dependent cofactor interaction properties of the estrogen receptors ERα and ERß, which form homo- or heterodimers whose distinctive regulatory properties are difficult to dissect using traditional methods. BRETFect uncovered the relative capacities of hetero- vs. homodimers to recruit receptor-specific cofactors and regulatory proteins, and to interact with common cofactors in the presence of receptor-specific ligands. BRETFect was also used to follow the assembly of ternary complexes between the V2R vasopressin receptor and two different intracellular effectors, illustrating its use for dissection of ternary protein-protein interactions engaged by G protein-coupled receptors. Our results indicate that BRETFect represents a powerful and versatile technique to monitor the dynamics of ternary interactions within multimeric complexes in live cells.


Subject(s)
Cytological Techniques/methods , Fluorescence Resonance Energy Transfer/methods , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Ternary Complex Factors/metabolism , HEK293 Cells , Humans , Luminescent Proteins , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/chemistry , Ternary Complex Factors/analysis , Ternary Complex Factors/chemistry
7.
J Am Heart Assoc ; 6(6)2017 Jun 25.
Article in English | MEDLINE | ID: mdl-28649088

ABSTRACT

BACKGROUND: Despite recent improvements in angioplasty and placement of drug-eluting stents in treatment of atherosclerosis, restenosis and in-stent thrombosis impede treatment efficacy and cause numerous deaths. Research efforts are needed to identify new molecular targets for blocking restenosis. We aim to establish angiogenic factor AGGF1 (angiogenic factor with G patch and FHA domains 1) as a novel target for blocking neointimal formation and restenosis after vascular injury. METHODS AND RESULTS: AGGF1 shows strong expression in carotid arteries; however, its expression is markedly decreased in arteries after vascular injury. AGGF1+/- mice show increased neointimal formation accompanied with increased proliferation of vascular smooth muscle cells (VSMCs) in carotid arteries after vascular injury. Importantly, AGGF1 protein therapy blocks neointimal formation after vascular injury by inhibiting the proliferation and promoting phenotypic switching of VSMCs to the contractile phenotype in mice in vivo. In vitro, AGGF1 significantly inhibits VSMCs proliferation and decreases the cell numbers at the S phase. AGGF1 also blocks platelet-derived growth factor-BB-induced proliferation, migration of VSMCs, increases expression of cyclin D, and decreases expression of p21 and p27. AGGF1 inhibits phenotypic switching of VSMCs to the synthetic phenotype by countering the inhibitory effect of platelet-derived growth factor-BB on SRF expression and the formation of the myocardin/SRF/CArG-box complex involved in activation of VSMCs markers. Finally, we show that AGGF1 inhibits platelet-derived growth factor-BB-induced phosphorylation of MEK1/2, ERK1/2, and Elk phosphorylation involved in the phenotypic switching of VSMCs, and that overexpression of Elk abolishes the effect of AGGF1. CONCLUSIONS: AGGF1 protein therapy is effective in blocking neointimal formation after vascular injury by regulating a novel AGGF1-MEK1/2-ERK1/2-Elk-myocardin-SRF/p27 signaling pathway.


Subject(s)
Angiogenic Proteins/administration & dosage , Carotid Artery Injuries/prevention & control , Carotid Stenosis/prevention & control , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Carotid Stenosis/genetics , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Cell Line , Cell Movement/drug effects , Cell Plasticity/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/metabolism , Phenotype , Phosphorylation , RNA Interference , Serum Response Factor/metabolism , Signal Transduction/drug effects , Ternary Complex Factors/metabolism , Trans-Activators/metabolism , Transfection
8.
Mol Cell ; 64(6): 1048-1061, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27867007

ABSTRACT

The ERK-regulated ternary complex factors (TCFs) act with the transcription factor serum response factor (SRF) to activate mitogen-induced transcription. However, the extent of their involvement in the immediate-early transcriptional response, and their wider functional significance, has remained unclear. We show that, in MEFs, TCF inactivation significantly inhibits over 60% of TPA-inducible gene transcription and impairs cell proliferation. Using integrated SRF ChIP-seq and Hi-C data, we identified over 700 TCF-dependent SRF direct target genes involved in signaling, transcription, and proliferation. These also include a significant number of cytoskeletal gene targets for the Rho-regulated myocardin-related transcription factor (MRTF) SRF cofactor family. The TCFs act as general antagonists of MRTF-dependent SRF target gene expression, competing directly with the MRTFs for access to SRF. As a result, TCF-deficient MEFs exhibit hypercontractile and pro-invasive behavior. Thus, competition between TCFs and MRTFs for SRF determines the balance between antagonistic proliferative and contractile programs of gene expression.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation , Serum Response Factor/genetics , Ternary Complex Factors/genetics , Animals , Base Sequence , Cell Line , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Profiling , Genetic Complementation Test , Humans , Mice , Serum Response Factor/metabolism , Signal Transduction , Ternary Complex Factors/antagonists & inhibitors , Ternary Complex Factors/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism
9.
Dev Biol ; 416(1): 173-186, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27235147

ABSTRACT

Elk proteins are Ets family transcription factors that regulate cell proliferation, survival, and differentiation in response to ERK (extracellular-signal regulated kinase)-mediated phosphorylation. Here we report the embryonic expression and function of Sp-Elk, the single Elk gene of the sea urchin Strongylocentrotus purpuratus. Sp-Elk is zygotically expressed throughout the embryo beginning at late cleavage stage, with peak expression occurring at blastula stage. Morpholino antisense-mediated knockdown of Sp-Elk causes blastula-stage developmental arrest and embryo disintegration due to apoptosis, a phenotype that is rescued by wild-type Elk mRNA. Development is also rescued by Elk mRNA encoding a serine to aspartic acid substitution (S402D) that mimics ERK-mediated phosphorylation of a conserved site that enhances DNA binding, but not by Elk mRNA encoding an alanine substitution at the same site (S402A). This demonstrates both that the apoptotic phenotype of the morphants is specifically caused by Elk depletion, and that phosphorylation of serine 402 of Sp-Elk is critical for its anti-apoptotic function. Knockdown of Sp-Elk results in under-expression of several regulatory genes involved in cell fate specification, cell cycle control, and survival signaling, including the transcriptional regulator Sp-Runt-1 and its target Sp-PKC1, both of which were shown previously to be required for cell survival during embryogenesis. Both Sp-Runt-1 and Sp-PKC1 have sequences upstream of their transcription start sites that specifically bind Sp-Elk. These results indicate that Sp-Elk is the signal-dependent activator of a feed-forward gene regulatory circuit, consisting also of Sp-Runt-1 and Sp-PKC1, which actively suppresses apoptosis in the early embryo.


Subject(s)
Cell Survival , Core Binding Factor alpha Subunits/metabolism , Sea Urchins/embryology , Signal Transduction , Ternary Complex Factors/metabolism , Animals , Apoptosis/genetics , Blastula , Cell Survival/genetics , Embryonic Development , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Oligonucleotides, Antisense , Phosphorylation , Promoter Regions, Genetic , Sea Urchins/genetics , Sea Urchins/metabolism , Signal Transduction/genetics
10.
Nat Commun ; 7: 11127, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27040916

ABSTRACT

Ternary complex (TC) and eIF4F complex assembly are the two major rate-limiting steps in translation initiation regulated by eIF2α phosphorylation and the mTOR/4E-BP pathway, respectively. How TC and eIF4F assembly are coordinated, however, remains largely unknown. We show that mTOR suppresses translation of mRNAs activated under short-term stress wherein TC recycling is attenuated by eIF2α phosphorylation. During acute nutrient or growth factor stimulation, mTORC1 induces eIF2ß phosphorylation and recruitment of NCK1 to eIF2, decreases eIF2α phosphorylation and bolsters TC recycling. Accordingly, eIF2ß mediates the effect of mTORC1 on protein synthesis and proliferation. In addition, we demonstrate a formerly undocumented role for CK2 in regulation of translation initiation, whereby CK2 stimulates phosphorylation of eIF2ß and simultaneously bolsters eIF4F complex assembly via the mTORC1/4E-BP pathway. These findings imply a previously unrecognized mode of translation regulation, whereby mTORC1 and CK2 coordinate TC and eIF4F complex assembly to stimulate cell proliferation.


Subject(s)
Casein Kinase II/physiology , Eukaryotic Initiation Factor-4F/metabolism , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Ternary Complex Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/physiology , Gene Expression Regulation , HEK293 Cells , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1 , Models, Genetic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Oncogene Proteins/metabolism , Peptide Chain Initiation, Translational , Phosphorylation , Signal Transduction , Stress, Physiological , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
11.
Br J Pharmacol ; 173(2): 305-18, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26493679

ABSTRACT

BACKGROUND AND PURPOSE: The rise in intracellular Ca(2+) stimulates the expression of the transcription factor c-Fos. Depending on the mode of entry of Ca(2+) into the cytosol, distinct signal transducers and transcription factors are required. Here, we have analysed the signalling pathway connecting a Ca(2+) influx via activation of transient receptor potential melastatin-3 (TRPM3) channels with enhanced c-Fos expression. EXPERIMENTAL APPROACH: Transcription of c-Fos promoter/reporter genes that were integrated into the chromatin via lentiviral gene transfer was analysed in HEK293 cells overexpressing TRPM3. The transcriptional activation potential of c-Fos was measured using a GAL4-c-Fos fusion protein. KEY RESULTS: The signalling pathway connecting TRPM3 stimulation with enhanced c-Fos expression requires the activation of MAP kinases. On the transcriptional level, three Ca(2+) -responsive elements, the cAMP-response element and the binding sites for the serum response factor (SRF) and AP-1, are essential for the TRPM3-mediated stimulation of the c-Fos promoter. Ternary complex factors are additionally involved in connecting TRPM3 stimulation with the up-regulation of c-Fos expression. Stimulation of TRPM3 channels also increases the transcriptional activation potential of c-Fos. CONCLUSIONS AND IMPLICATIONS: Signalling molecules involved in connecting TRPM3 with the c-Fos gene are MAP kinases and the transcription factors CREB, SRF, AP-1 and ternary complex factors. As c-Fos constitutes, together with other basic region leucine zipper transcription factors, the AP-1 transcription factor complex, the results of this study explain TRPM3-induced activation of AP-1 and connects TRPM3 with the biological functions regulated by AP-1.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Genes, fos/physiology , Mitogen-Activated Protein Kinases/metabolism , TRPM Cation Channels/metabolism , Ternary Complex Factors/metabolism , Transcription Factor AP-1/metabolism , Gene Expression Regulation , Genes, fos/drug effects , HEK293 Cells , Humans , Pregnenolone/pharmacology
12.
Mol Pharmacol ; 87(4): 617-28, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25576487

ABSTRACT

The steroid pregnenolone sulfate activates the transcription factor activator protein-1 (AP-1) via stimulation of transient receptor potential melastatin-3 (TRPM3) channels. Here, we show that the signaling pathway requires an influx of Ca(2+) ions into the cells and a rise in the intracellular Ca(2+) levels. The upregulation of AP-1 was attenuated in cells that overexpressed mitogen activated protein kinase phosphatase-1, indicating that Ca(2+) ions prolong the signaling cascade via activation of mitogen activated protein kinases. On the transcriptional level, expression of a dominant-negative mutant of the basic region leucine zipper protein c-Jun, a major constituent of the AP-1 transcription factor complex, or expression of a c-Jun-specific short hairpin RNA attenuated pregnenolone sulfate-induced AP-1 activation. In addition, stimulation of TRPM3 channels increased the transcriptional activation potential of the basic region leucine zipper protein ATF2. Inhibition of ATF2 target gene expression via expression of a dominant-negative mutant of ATF2 or expression of an ATF2-specific short hairpin RNA interfered with TRPM3-mediated stimulation of AP-1. Moreover, we show that a dominant-negative mutant of the ternary complex factor (TCF) Elk-1 attenuated the upregulation of AP-1 following stimulation of TRPM3 channels. Thus, c-Jun, ATF2, and TCFs are required to connect the intracellular signaling cascade elicited by activation of TRPM3 channels with enhanced transcription of AP-1-regulated genes. We conclude that pregnenolone sulfate-induced TRPM3 channel activation changes the gene expression pattern of the cells by activating transcription of c-Jun-, ATF2-, and TCF-controlled genes.


Subject(s)
Activating Transcription Factor 2/metabolism , Calcium/metabolism , Proto-Oncogene Proteins c-jun/metabolism , TRPM Cation Channels/metabolism , Ternary Complex Factors/metabolism , Transcription Factor AP-1/metabolism , Activating Transcription Factor 2/genetics , Cations , HEK293 Cells , Humans , Pregnenolone/pharmacology , Proto-Oncogene Proteins c-jun/genetics , TRPM Cation Channels/genetics , Ternary Complex Factors/genetics , Transcription, Genetic , Up-Regulation , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism
13.
J Clin Endocrinol Metab ; 99(10): E1988-96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24926947

ABSTRACT

BACKGROUND: IGF-I is mainly sequestered in a 150-kDa ternary complex with IGF binding protein (IGFBP)-3 and the acid-labile subunit. Data on complex formation and factors influencing formation have not been established. Dissociation of IGF-I from the ternary complex is in part regulated by proteolysis of IGFBP-3, which reduces its affinity for IGF-I. Short small for gestational age (SGA) children have lower IGF-I and IGFBP-3 levels compared with healthy peers. OBJECTIVE: The objective of the study was to determine complex formation in healthy normal-statured children and assess variables influencing complex formation. Second, we determined complex formation in short SGA children. DESIGN/METHODS: Complex formation was assessed using (125)I-hIGF-I column chromatography in 70 controls (40 boys), median age 10.6 years, and 40 short SGA children (25 boys), median age 8.6 years. IGFBP-3 was determined by Western immunoblotting. RESULTS: (125)I-hIGF-I complex formation showed an age-specific pattern in healthy controls. Variables positively influencing ternary complex formation were higher serum IGF-I levels compared with IGFBP-3 levels (P < .001) and lower serum IGF-II (P < .001) and IGFBP-1 levels (P < .001). In addition, a higher presence of proteolyzed IGFBP-3 negatively influenced 150-kDa complex formation (P = .006). At a young age, healthy children showed considerable IGFBP-3 proteolytic activity, which declined with aging (P < .001). IGFBP-3 proteolytic activity was negatively correlated with IGF-I levels (P < .001). Compared with healthy controls, short SGA children showed reduced IGF-I levels (-1.3 vs 0.1 SD score) and increased proteolyzed IGFBP-3 (35.1% vs 12.2%). CONCLUSION: Age-specific normative values for (125)I-hIGF-I 150-kDa ternary complex formation are presented. A decrease in IGF-I and an increase in IGF-II, IGFBP-1, and IGFBP-3 proteolytic activity associate with reduced (125)I-hIGF-I ternary complex formation. Our results suggest that in conditions in which IGF-I levels are low, such as young age and in short SGA children, IGFBP-3 proteolytic activity is increased to ensure IGF-I bioavailability.


Subject(s)
Adolescent Development/physiology , Child Development/physiology , Infant, Small for Gestational Age/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Ternary Complex Factors/metabolism , Adolescent , Age Factors , Child , Child, Preschool , Chromatography/methods , Female , Humans , Infant , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , Iodine Radioisotopes , Male , Young Adult
14.
Autophagy ; 9(10): 1467-74, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23939028

ABSTRACT

Atg17, in complex with Atg29 and Atg31, constitutes a key module of the Atg1 kinase signaling complex and functions as an important organizer of the phagophore assembly site in the yeast Saccharomyces cerevisiae. We have determined the three-dimensional reconstruction of the full S. cerevisiae Atg17-Atg31-Atg29 complex by single-particle electron microscopy. Our structure shows that Atg17-Atg31-Atg29 is dimeric and adopts a relatively rigid and extended "S-shape" architecture with an end-to-end distance of approximately 345 Å. Subunit mapping analysis indicated that Atg17 mediates dimerization and generates a central rod-like scaffold, while Atg31 and Atg29 form two globular domains that are tethered to the concave sides of the scaffold at the terminal regions. Finally, our observation that Atg17 adopts multiple conformations in the absence of Atg31 and Atg29 suggests that the two smaller components play key roles in defining and maintaining the distinct curvature of the ternary complex.


Subject(s)
Autophagy/physiology , Carrier Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Autophagy-Related Proteins , Microscopy, Electron/methods , Saccharomyces cerevisiae/cytology , Ternary Complex Factors/metabolism
15.
PLoS One ; 8(3): e58756, 2013.
Article in English | MEDLINE | ID: mdl-23505557

ABSTRACT

Primate-specific Mas-related G protein-coupled receptors-X1 (MRGPR-X1) are highly enriched in dorsal root ganglia (DRG) neurons and induce acute pain. Herein, we analyzed effects of MRGPR-X1 on serum response factors (SRF) or nuclear factors of activated T cells (NFAT), which control expression of various markers of chronic pain. Using HEK293, DRG neuron-derived F11 cells and cultured rat DRG neurons recombinantly expressing human MRGPR-X1, we found activation of a SRF reporter gene construct and induction of the early growth response protein-1 via extracellular signal-regulated kinases-1/2 known to play a significant role in the development of inflammatory pain. Furthermore, we observed MRGPR-X1-induced up-regulation of the chemokine receptor 2 (CCR2) via NFAT, which is considered as a key event in the onset of neuropathic pain and, so far, has not yet been described for any endogenous neuropeptide. Up-regulation of CCR2 is often associated with increased release of its endogenous agonist chemokine ligand 2 (CCL2). We also found MRGPR-X1-promoted release of CCL2 in a human connective tissue mast cell line endogenously expressing MRGPR-X1. Thus, we provide first evidence to suggest that MRGPR-X1 induce expression of chronic pain markers in DRG neurons and propose a so far unidentified signaling circuit that enhances chemokine signaling by acting on two distinct yet functionally co-operating cell types. Given the important role of chemokine signaling in pain chronification, we propose that interruption of this signaling circuit might be a promising new strategy to alleviate chemokine-promoted pain.


Subject(s)
Chemokine CCL2/metabolism , Ganglia, Spinal/metabolism , Mast Cells/metabolism , Receptors, CCR2/genetics , Receptors, G-Protein-Coupled/metabolism , Sensory Receptor Cells/metabolism , Animals , Bradykinin/pharmacology , Calcineurin/metabolism , Calcium/metabolism , Cell Line , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Genes, fos , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NFATC Transcription Factors/metabolism , Peptide Fragments/pharmacology , Rats , Receptors, CCR2/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Serum Response Factor/metabolism , Ternary Complex Factors/metabolism
16.
FEBS J ; 280(10): 2207-15, 2013 May.
Article in English | MEDLINE | ID: mdl-23375101

ABSTRACT

The syndecans are a family of heparan sulfate-decorated cell-surface proteoglycans: matrix receptors with roles in cell adhesion and growth factor signaling. Their heparan sulfate chains recognize 'heparin-binding' motifs that are ubiquitously present in the extracellular matrix, providing the means for syndecans to constitutively bind and cluster to sites of cell-matrix adhesion. Emerging evidence suggests that specialized docking sites in the syndecan extracellular domains may serve to localize other receptors to these sites as well, including integrins and growth factor receptor tyrosine kinases. A prototype of this mechanism is capture of the αvß3 integrin and insulin-like growth factor 1 receptor (IGF1R) by syndecan-1 (Sdc1), forming a ternary receptor complex in which signaling downstream of IGF1R activates the integrin. This Sdc1-coupled ternary receptor complex is especially prevalent on tumor cells and activated endothelial cells undergoing angiogenesis, reflecting the up-regulated expression of αvß3 integrin in such cells. As such, much effort has focused on developing therapeutic agents that target this integrin in various cancers. Along these lines, the site in the Sdc1 ectodomain that is responsible for capture and activation of the αvß3 or αvß5 integrins by IGF1R can be mimicked by a short peptide called 'synstatin', which competitively displaces the integrin and IGF1R kinase from the syndecan and inactivates the complex. This review summarizes our current knowledge of the Sdc1-coupled ternary receptor complex and the efficacy of synstatin as an emerging therapeutic agent to target this signaling mechanism.


Subject(s)
Cell Transformation, Neoplastic , Integrin alphaVbeta3/metabolism , Neovascularization, Pathologic/metabolism , Receptor, IGF Type 1/metabolism , Syndecan-1/metabolism , Ternary Complex Factors/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Adhesion , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Mice , Neovascularization, Pathologic/pathology , Peptides/pharmacology , Protein Binding , Recombinant Proteins/pharmacology , Syndecan-1/antagonists & inhibitors
17.
FEBS J ; 280(10): 2194-206, 2013 May.
Article in English | MEDLINE | ID: mdl-23331867

ABSTRACT

Vascular endothelial growth factor (VEGF)-stimulated angiogenesis depends on a cross-talk mechanism involving VEGF receptor 2 (VEGFR2), vascular endothelial (VE)-cadherin and the αVß3 integrin. Because we have shown that αVß3 integrin activation is dependent on its incorporation, along with the insulin-like growth factor-1 receptor (IGF1R) kinase, into a ternary receptor complex organized by the matrix receptor syndecan-1 (Sdc1), we questioned the role of this core complex in VEGF-stimulated angiogenesis. We find that the Sdc1-coupled ternary receptor complex is required for VEGF signalling and for stimulation of vascular endothelial cell migration by vascular endothelial cadherin (VE-cadherin) engagement. VE-cadherin binding to Fc/VE-cadherin extracellular domain chimera activates Sdc1-coupled IGF1R and αvß3 integrin; this depends on VEGFR2 and c-Src activated by the cadherin. Blocking homotypic VE-cadherin engagement disrupts VEGF-stimulated cell migration, which is restored by clustering the cadherin in the absence of cell-cell adhesion. This cadherin-dependent stimulation requires VEGFR2 and IGF1R and is blocked by synstatin (SSTN)(92-119), a peptide that competitively disrupts the Sdc1-coupled ternary complex and prevents the αVß3 integrin activation required for VEGFR2 activation. VEGFR2-stimulated angiogenesis in the mouse aortic ring explant assay is disrupted by SSTN, although only early in the process, suggesting that IGF1R coupling to Sdc1 and αVß3 integrin comprises a core activation mechanism activated by VE-cadherin that is necessary for VEGFR2 and integrin activation in the initial stages of endothelial cell dissemination during angiogenesis.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/metabolism , Integrin alphaVbeta3/metabolism , Receptors, Somatomedin/metabolism , Syndecan-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Antibodies/metabolism , Aorta/drug effects , Aorta/metabolism , Cadherins/antagonists & inhibitors , Cell Adhesion , Cell Movement , Cells, Cultured , Collagen/metabolism , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neovascularization, Physiologic/drug effects , Peptides/pharmacology , Phosphorylation , Protein Binding , Protein Interaction Mapping , Receptor Cross-Talk , Receptors, Somatomedin/antagonists & inhibitors , Ternary Complex Factors/metabolism , Vascular Endothelial Growth Factor A/pharmacology
18.
Biochim Biophys Acta ; 1829(3-4): 306-17, 2013.
Article in English | MEDLINE | ID: mdl-23092677

ABSTRACT

The synthesis of ribosomal RNA (rRNA) precursor molecules by RNA polymerase I (Pol I) terminates with the dissociation of the protein-DNA-RNA ternary complex. Based on in vitro results the mechanism of Pol I termination appeared initially to be rather conserved and simple until this process was more thoroughly re-investigated in vivo. A picture emerged that Pol I termination seems to be connected to co-transcriptional processing, re-initiation of transcription and, possibly, other processes downstream of Pol I transcription units. In this article, our current understanding of the mechanism of Pol I termination and how this process might be implicated in other biological processes in yeast and mammals is summarized and discussed. This article is part of a Special Issue entitled: Transcription by Odd Pols.


Subject(s)
RNA Polymerase I/metabolism , Transcription Termination, Genetic , Animals , Humans , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Precursors/biosynthesis , RNA Precursors/genetics , RNA, Ribosomal/biosynthesis , RNA, Ribosomal/genetics , Ternary Complex Factors/metabolism , Yeasts/genetics , Yeasts/metabolism
19.
Curr Biol ; 22(21): 2017-26, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23063436

ABSTRACT

BACKGROUND: The ternary complex factors (TCFs; Elk1, Net, and Sap-1) are growth factor-responsive transcription cofactors of serum response factor (SRF) and are activated by MAP kinase (MAPK) phosphorylation to regulate immediate early gene transcription. Although cell adhesion also can regulate immediate early genes and proliferation, the mechanism for this effect has remained unexplored. RESULTS: Restricting adhesion and spreading of G(0)-synchronized cells on substrates with decreasing size of micropatterned islands of fibronectin suppressed serum-induced immediate early gene expression and S phase entry. Knockdown of Sap-1 decreased expression of the immediate early genes egr1 and fos and subsequent proliferation normally present with high adhesion, whereas knockdown of Net rescued egr1 and fos expression and proliferation normally suppressed by low adhesion. Chromatin immunoprecipitation studies showed increased occupancy of egr1 and fos promoters by Sap-1 with high adhesion, whereas low adhesion increased Net occupancy. This switch in TCF promoter binding was regulated by an adhesion-mediated switch in MAPK activity. Increasing adhesion enhanced serum-induced JNK activity while suppressing p38 activity, leading to increased Sap-1 phosphorylation and Net dephosphorylation, and switching Net with Sap-1 at egr1 and fos promoters to support proliferation. Microarray studies confirmed this switch in TCF regulation of proliferative genes and uncovered novel gene targets and functions coregulated by Sap-1 and Net. CONCLUSIONS: These data demonstrate a key role for the TCFs in adhesion-induced transcription and proliferation and reveal a novel MAPK/TCF transcriptional switch that controls this process.


Subject(s)
Cell Adhesion/physiology , Mitogen-Activated Protein Kinases/metabolism , Serum Response Factor/metabolism , Ternary Complex Factors/metabolism , Transcription, Genetic , 3T3 Cells , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Cell Proliferation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Sodium Channels/genetics , Sodium Channels/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Genes Cells ; 17(8): 688-97, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22734669

ABSTRACT

Dock3, a new member of the guanine nucleotide exchange factor family, causes cellular morphological changes by activating the small GTPase Rac1. Overexpression of Dock3 in neural cells promotes neurite outgrowth through the formation of a protein complex with Fyn and WAVE downstream of brain-derived neurotrophic factor (BDNF) signaling. Here, we report a novel Dock3-mediated BDNF pathway for neurite outgrowth. We show that Dock3 forms a complex with Elmo and activated RhoG downstream of BDNF-TrkB signaling and induces neurite outgrowth via Rac1 activation in PC12 cells. We also show the importance of Dock3 phosphorylation in Rac1 activation and show two key events that are necessary for efficient Dock3 phosphorylation: membrane recruitment of Dock3 and interaction of Dock3 with Elmo. These results suggest that Dock3 plays important roles downstream of BDNF signaling in the central nervous system where it stimulates actin polymerization by multiple pathways.


Subject(s)
Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Receptor, trkB/metabolism , Signal Transduction , Ternary Complex Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , COS Cells , Carrier Proteins/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Chlorocebus aethiops , Enzyme Activation , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Exchange Factors , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurites/drug effects , Neurites/physiology , Neuropeptides/genetics , Neuropeptides/metabolism , PC12 Cells , Phosphorylation , Protein Interaction Mapping , Protein Transport , Rats , Receptor, trkB/genetics , Ternary Complex Factors/genetics , Transfection , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein , rho GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL