Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
CNS Neurosci Ther ; 30(2): e14585, 2024 02.
Article in English | MEDLINE | ID: mdl-38421133

ABSTRACT

INTRODUCTION: Serum response factor (SRF) is important in muscle development, tissue repair, and neuronal regulation. OBJECTIVES: This research aims to thoroughly examine the effects of SRF on spinal cord injury (SCI) and its ability to significantly impact the recovery and regeneration of neuronal axons. METHODS: The researchers created rat models of SCI and scratch injury to primary spinal cord neurons to observe the expression of relevant factors after neuronal injury. RESULTS: We found that the SRF, Ras, Raf, and cofilin levels increased after injury and gradually returned to normal levels. Afterward, researchers gave rats with SCI an SRF inhibitor (CCG1423) and studied the effects with nuclear magnetic resonance and transmission electron microscopy. The SRF inhibitor rodents had worse spinal cord recovery and axon regrowth than the control group. And the apoptosis of primary neurons after scratch injury was significantly higher in the SRF inhibitor group. Additionally, the researchers utilized lentiviral transfection to modify the SRF expression in neurons. SRF overexpression increased neuron migration while silencing SRF decreased it. Finally, Western blotting and RT-PCR were conducted to examine the expression changes of related factors upon altering SRF expression. The results revealed SRF overexpression increased Ras, Raf, and cofilin expression. Silencing SRF decreased Ras, Raf, and Cofilin expression. CONCLUSION: Based on our research, the SRF promotes axonal regeneration by activating the "Ras-Raf-Cofilin" signaling pathway.


Subject(s)
Actin Depolymerizing Factors , Spinal Cord Injuries , Rats , Animals , Actin Depolymerizing Factors/metabolism , Actin Depolymerizing Factors/pharmacology , Serum Response Factor/genetics , Serum Response Factor/metabolism , Serum Response Factor/pharmacology , Spinal Cord Injuries/pathology , Neurons/metabolism , Axons , Spinal Cord/metabolism , Signal Transduction , Nerve Regeneration , Recovery of Function/physiology
2.
Mediators Inflamm ; 2022: 1818758, 2022.
Article in English | MEDLINE | ID: mdl-36248188

ABSTRACT

Lysophosphatidic acid (LPA) has disruptive effects on lumbar spinal stenosis (LSS). Recently, LPA has been reported to be involved in spinal cord neuronal injury and toxicity, promoting the pathogenesis of LSS. However, the exact effects of LPA on spinal cord neurons remain unknown. The purpose of this study is to investigate the effects of LPA (18 : 1) on spinal cord neuronal cytotoxicity, apoptosis, DNA damage, and oxidative stress. After clinical detection of LPA secretion, spinal cord neurons were treated with LPA (18 : 1); cell viability was analyzed by MTT assay, and LDH leakage was detected by LDH kit; cell apoptosis was detected by flow cytometry; ROS production was measured by DCFDA staining and MitoSOX Red Staining; the activation of the Gα12/Gα13 signaling pathway was detected by serum response factor response element (SRF-RE) luciferase reporter gene; the relationship among LPA, LPA4/6, and ROCK was examined by western blotting. In spinal cord neurons treated with LPA (18 : 1), cellular activity decreased and LDH release increased. The Rho kinase inhibitor (Y-27632) can attenuate LPA-induced apoptosis, DNA damage, and oxidative stress in spinal cord neurons. Moreover mechanistic investigation indicated that LPA (18 : 1) activates Gα12/13-Rho-ROCK2-induced apoptosis, DNA damage, and oxidative stress in spinal cord neurons by upregulating LPA4/LPA6 receptors. Further, the Rho kinase inhibitor Y-27632 attenuates the effects of LPA by downregulating LPA4/LPA6 receptors. Taken together, the possible mechanism by which LPA secretion in LSS patients aggravates patient injury was further elucidated using an LPA-induced spinal cord neuronal injury cell model in vitro.


Subject(s)
Receptors, Lysophosphatidic Acid , Spinal Cord Injuries , Amides , Apoptosis , DNA Damage , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/pharmacology , Humans , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Neurons/metabolism , Oxidative Stress , Pyridines , Reactive Oxygen Species/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Purinergic P2/metabolism , Serum Response Factor/metabolism , Serum Response Factor/pharmacology , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , rho-Associated Kinases/metabolism , rho-Associated Kinases/pharmacology
3.
J Pineal Res ; 73(4): e12833, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36106819

ABSTRACT

In vitro differentiation of stem cells into functional gametes remains of great interest in the biomedical field. Skin-derived stem cells (SDSCs) are an adult stem cells that provides a wide range of clinical applications without inherent ethical restrictions. In this paper, porcine SDSCs were successfully differentiated into primordial germ cell-like cells (PGCLCs) in conditioned media. The PGCLCs were characterized in terms of cell morphology, marker gene expression, and epigenetic properties. Furthermore, we also found that 25 µM melatonin (MLT) significantly increased the proliferation of the SDSC-derived PGCLCs while acting through the MLT receptor type 1 (MT1). RNA-seq results found the mitogen-activated protein kinase (MAPK) signaling pathway was more active when PGCLCs were cultured with MLT. Moreover, the effect of MLT was attenuated by the use of S26131 (MT1 antagonist), crenolanib (platelet-derived growth factor receptor inhibitor), U0126 (mitogen-activated protein kinase kinase inhibitor), or CCG-1423 (serum response factor transcription inhibitor), suggesting that MLT promotes the proliferation processes through the MAPK pathway. Taken together, this study highlights the role of MLT in promoting PGCLCs proliferation. Importantly, this study provides a suitable in vitro model for use in translational studies and could help to answer numerous remaining questions related to germ cell physiology.


Subject(s)
Melatonin , Swine , Animals , Melatonin/pharmacology , Melatonin/metabolism , Serum Response Factor/metabolism , Serum Response Factor/pharmacology , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Germ Cells/metabolism , Stem Cells , Cell Differentiation , Cell Proliferation , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/pharmacology
4.
J Cardiovasc Pharmacol ; 80(4): 574-582, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35881905

ABSTRACT

ABSTRACT: Inositol 1, 4, 5-trisphosphate (IP3) signaling-mediated calcium release drives the contraction of vascular smooth muscles and hence regulates blood vessel volume and blood pressure. Melatonin supplementation has been suggested to be beneficial for hypertension. To determine whether the blood pressure-lowering effect of melatonin was accounted for by IP3 signaling, we evaluated the vasoconstriction response and IP3 signaling in isolated mouse thoracic aortic rings during melatonin incubation. C57BL/6 mice were given intraperitoneal injections daily with melatonin, and the systolic blood pressure and contractility of aortic rings from melatonin-treated mice were decreased, and the contraction suppression effect of melatonin was attributed to the impaired expression of contractile proteins in vascular smooth muscle cells rather than IP3 signaling. Our results further showed that melatonin increased the expression of γ-secretase, which could cleave and release the notch intracellular domain, and the notch intracellular domain prevented the transcription of contractile genes by interfering with the interaction between serum response factor and myocardin, the master regulator of contractile protein. In this article, we report a novel mechanism by which melatonin regulates smooth muscle contractility that does not depend on IP3 signaling.


Subject(s)
Melatonin , Vasoconstriction , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/pharmacology , Animals , Calcium/metabolism , Contractile Proteins/metabolism , Contractile Proteins/pharmacology , Inositol/metabolism , Inositol/pharmacology , Melatonin/pharmacology , Mice , Mice, Inbred C57BL , Muscle Contraction , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins , Serum Response Factor/metabolism , Serum Response Factor/pharmacology , Trans-Activators
6.
J Neurosci ; 33(48): 18836-48, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24285890

ABSTRACT

Axonal injury generates growth inert retraction bulbs with dynamic cytoskeletal properties that are severely compromised. Conversion of "frozen" retraction bulbs into actively progressing growth cones is a major aim in axon regeneration. Here we report that murine serum response factor (SRF), a gene regulator linked to the actin cytoskeleton, modulates growth cone actin dynamics during axon regeneration. In regeneration-competent facial motoneurons, Srf deletion inhibited axonal regeneration. In wild-type mice after nerve injury, SRF translocated from the nucleus to the cytoplasm, suggesting a cytoplasmic SRF function in axonal regeneration. Indeed, adenoviral overexpression of cytoplasmic SRF (SRF-ΔNLS-GFP) stimulated axonal sprouting and facial nerve regeneration in vivo. In primary central and peripheral neurons, SRF-ΔNLS-GFP stimulated neurite outgrowth, branch formation, and growth cone morphology. Furthermore, we uncovered a link between SRF and the actin-severing factor cofilin during axonal regeneration in vivo. Facial nerve axotomy increased the total cofilin abundance and also nuclear localization of phosphorylated cofilin in a subpopulation of lesioned motoneurons. This cytoplasmic-to-nucleus translocation of P-cofilin upon axotomy was reduced in motoneurons expressing SRF-ΔNLS-GFP. Finally, we demonstrate that cytoplasmic SRF and cofilin formed a reciprocal regulatory unit. Overexpression of cytoplasmic SRF reduced cofilin phosphorylation and vice versa: overexpression of cofilin inhibited SRF phosphorylation. Therefore, a regulatory loop consisting of SRF and cofilin might take part in reactivating actin dynamics in growth-inert retraction bulbs and facilitating axon regeneration.


Subject(s)
Actin Depolymerizing Factors/physiology , Axons/drug effects , Cytoplasm/metabolism , Nerve Regeneration/drug effects , Serum Response Factor/pharmacology , Actins/metabolism , Animals , Axotomy , Cytoplasm/drug effects , Facial Nerve/physiology , Female , Green Fluorescent Proteins , Male , Mice , Peripheral Nerves/cytology , Peripheral Nerves/drug effects , Phosphorylation , Polymerase Chain Reaction , Subcellular Fractions/metabolism
7.
Cell Signal ; 25(3): 630-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23195829

ABSTRACT

RhoGEF17, the product of the ARHGEF17 gene, is a Rho-specific guanine nucleotide exchange factor (GEF) with an unusual structure and so far unknown function. In order to get insights in its regulation, we studied a variety of signaling pathways for activation of recombinantly expressed RhoGEF17. We found that in the presence of stable cGMP analogs RhoGEF17 associates with and is phosphorylated by co-expressed cGKIα at distinct phosphorylation sites leading to a cooperative activation of RhoA, the Rho dependent kinases (ROCK) and serum response factor-induced gene transcription. Activation of protein kinase A did not induce phosphorylation of RhoGEF17 nor altered its activity. Furthermore, we obtained evidence for a ROCK-driven positive feedback mechanism involving serine/threonine protein phosphatases, which further enhanced cGMP/cGKIα-induced RhoGEF17 activation. By using mutants of RhoA which are phosphorylation resistant to cGK or mimic phosphorylation at serine 188, we could show that RhoGEF17 is able to activate RhoA independently of its phosphorylation state. Together with the ROCK-enforced activation of RhoGEF17 by cGMP/cGKIα, this might explain why expression of RhoGEF17 switches the inhibitory effect of cGMP/cGKIα on serum-induced RhoA activation into a stimulatory one. We conclude that RhoGEF17, depending on its expression profile and level, might drastically alter the effect of cGMP/cGK involving signaling pathways on RhoA-activated downstream effectors.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Cell Line , Cyclic GMP/pharmacology , Enzyme Activation/drug effects , Guanine Nucleotide Exchange Factors/genetics , Humans , Mutagenesis, Site-Directed , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Rho Guanine Nucleotide Exchange Factors , Serum Response Factor/pharmacology , Signal Transduction , Transcription, Genetic/drug effects , rhoA GTP-Binding Protein/metabolism
8.
PLoS One ; 6(10): e26089, 2011.
Article in English | MEDLINE | ID: mdl-22022520

ABSTRACT

During brain development, growth cones respond to attractive and repulsive axon guidance cues. How growth cones integrate guidance instructions is poorly understood. Here, we demonstrate a link between BDNF (brain derived neurotrophic factor), promoting axonal branching and ephrin-A5, mediating axonal repulsion via Eph receptor tyrosine kinase activation. BDNF enhanced growth cone filopodial dynamics and neurite branching of primary neurons. We show that ephrin-A5 antagonized this BDNF-evoked neuronal motility. BDNF increased ERK phosphorylation (P-ERK) and nuclear ERK entry. Ephrin-A5 suppressed BDNF-induced ERK activity and might sequester P-ERK in the cytoplasm. Neurotrophins are well established stimulators of a neuronal immediate early gene (IEG) response. This is confirmed in this study by e.g. c-fos, Egr1 and Arc upregulation upon BDNF application. This BDNF-evoked IEG response required the transcription factor SRF (serum response factor). Notably, ephrin-A5 suppressed a BDNF-evoked neuronal IEG response, suggesting a role of Eph receptors in modulating gene expression. In opposite to IEGs, long-term ephrin-A5 application induced cytoskeletal gene expression of tropomyosin and actinin. To uncover specific Eph receptors mediating ephrin-As impact on neurotrophin signaling, EphA7 deficient mice were analyzed. In EphA7 deficient neurons alterations in growth cone morphology were observed. However, ephrin-A5 still counteracted neurotrophin signaling suggesting that EphA7 is not required for ephrin and BDNF crosstalk. In sum, our data suggest an interaction of ephrin-As and neurotrophin signaling pathways converging at ERK signaling and nuclear gene activity. As ephrins are involved in development and function of many organs, such modulation of receptor tyrosine kinase signaling and gene expression by Ephs might not be limited to the nervous system.


Subject(s)
Cell Movement/drug effects , Ephrin-A5/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Nerve Growth Factors/pharmacology , Neurons/cytology , Neurons/enzymology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Communication/drug effects , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Enzyme Activation/drug effects , Genes, Immediate-Early/genetics , Growth Cones/drug effects , Mice , Neurites/drug effects , Neurites/enzymology , Neurons/drug effects , Phosphorylation/drug effects , Protein Transport/drug effects , Receptors, Eph Family/metabolism , Serum Response Factor/pharmacology , Signal Transduction/drug effects
9.
Arterioscler Thromb Vasc Biol ; 24(9): 1602-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15242862

ABSTRACT

OBJECTIVE: Hex (hematopoietically expressed homeobox), a member of homeobox family of transcription factors, has been implicated in the vascular development because of its expression in hemangioblast, a hypothetical stem cell that gives rise to both angioblasts and hematopoietic lineages. In the present study, we examined the role of Hex in the differentiation of vascular smooth muscle cells. METHODS AND RESULTS: We constructed adenovirus expressing Hex, to which we refer to as AxCA/Hex, and transduced murine embryonic fibroblasts, 10T1/2 cells. Northern blot analyses showed that Hex increased the mRNA levels of smooth muscle alpha-actin and SM22alpha but not of calponin and smooth muscle myosin heavy chain. Transient transfection assays showed that Hex activates the transcription from the SM22alpha promoter in a CArG box-dependent manner. Electrophoretic mobility shift assays demonstrate that Hex is not able to bind to CArG box, but binding of serum responsive factor (SRF) to CArG box is enhanced in AxCA/Hex-transduced cells. Recombinant Hex protein produced by in vitro translation system augmented the binding activity of SRF to CArG box. Immunoprecipitation experiments revealed the physical association between Hex and SRF. CONCLUSIONS: Hex induces transcription of the SM22alpha gene by facilitating the interaction between SRF and its cognate binding site in pluripotent embryonic fibroblasts. This study demonstrates that Hex, a hematopoietically expressed homeobox protein, induces transcription of the SM22alpha gene by facilitating the interaction between SRF and its cognate binding site in embryonic fibroblasts. These findings will provide the clue for understanding the mechanisms by which bone marrow-derived SMC precursor cells undergo differentiation.


Subject(s)
Actins/biosynthesis , Gene Expression Regulation/physiology , Homeodomain Proteins/physiology , Microfilament Proteins/biosynthesis , Muscle Proteins/biosynthesis , Serum Response Element/genetics , Serum Response Factor/pharmacology , Actins/genetics , Animals , Binding Sites , Cells, Cultured/metabolism , Electrophoretic Mobility Shift Assay , Fibroblasts/metabolism , Genes, Reporter , Mice , Microfilament Proteins/genetics , Muscle Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Protein Interaction Mapping , Recombinant Fusion Proteins/physiology , Serum Response Factor/metabolism , Transcription Factors
10.
Arterioscler Thromb Vasc Biol ; 24(8): 1384-90, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15217807

ABSTRACT

OBJECTIVE: Transforming growth factor-beta1 (TGFbeta1) and fibroblast growth factor (FGF) families play a pivotal role during vascular development and in the pathogenesis of vascular disease. However, the interaction of intracellular signaling evoked by each of these growth factors is not well understood. The present study was undertaken to examine the molecular mechanisms that mediate the effects of TGFbeta1 and basic FGF (bFGF) on smooth muscle cell (SMC) gene expression. METHODS AND RESULTS: TGFbeta1 induction of SMC gene expression, including smooth muscle protein 22-alpha (SM22alpha) and smooth muscle alpha-actin, was examined in the pluripotent 10T1/2 cells. Marked increase in these mRNA levels by TGFbeta1 was inhibited by c-Src-tyrosine kinase inhibitors and protein synthesis inhibitor cycloheximide. Functional studies with deletion and site-directed mutation analysis of the SM22alpha promoter demonstrated that TGFbeta1 activated the SM22alpha promoter through a CC(A/T-rich)6GG (CArG) box, which serves as a serum response factor (SRF)-binding site. TGFbeta1 increased SRF expression through an increase in transcription of the SRF gene. In the presence of bFGF, TGFbeta1 induction of SMC marker gene expression was significantly attenuated. Transient transfection assays showed that bFGF significantly suppressed induction of the SM22alpha promoter-driven luciferase activity by TGFbeta1, whereas bFGF had no effects on the TGFbeta1-mediated increase in SRF expression and SRF:DNA binding activity. Mitogen-activated protein kinase kinase-1 (MEK1) inhibitor PD98059 abrogated the bFGF-mediated suppression of TGFbeta1-induced SMC gene expression. CONCLUSIONS: Our data suggest that bFGF-induced MEK/extracellular signal-regulated kinase signaling plays an antagonistic role in TGFbeta1-induced SMC gene expression through suppression of the SRF function. These data indicate that opposing effects of bFGF and TGFbeta1 on SMC gene expression control the phenotypic plasticity of SMCs.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , src-Family Kinases/physiology , Actins/biosynthesis , Actins/genetics , Animals , Aorta , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Cell Differentiation/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cycloheximide/pharmacology , DNA/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/physiology , Mice , Mice, Inbred C3H , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Mutagenesis, Site-Directed , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic/drug effects , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Recombinant Proteins/pharmacology , Serum Response Element/drug effects , Serum Response Factor/metabolism , Serum Response Factor/pharmacology , Transcription, Genetic/drug effects , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , src-Family Kinases/antagonists & inhibitors , Calponins
11.
J Biol Chem ; 278(33): 30642-51, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12788937

ABSTRACT

The chaperonin-containing t-complex polypeptide 1 (CCT) is a molecular chaperone that facilitates protein folding in eukaryotic cytosol, and the expression of CCT is highly dependent on cell growth. We show here that transcription of the gene encoding the theta subunit of mouse CCT, Cctq, is regulated by the ternary complex factors (TCFs), Elk-1, Sap-1a, and Net (Sap-2). Reporter gene assay using HeLa cells indicated that the Cctq gene promoter contains a cis-acting element of the CCGGAAGT sequence (CQE1) at -36 bp. The major CQE1-binding proteins in HeLa cell nuclear extract was recognized by anti-Elk-1 or anti-Sap-1a antibodies in electrophoretic mobility shift assay, and recombinant Elk-1, Sap-1a, or Net specifically recognized CQE1. The CQE1-dependent transcriptional activity in HeLa cells was virtually abolished by overexpression of the DNA binding domains of TCFs. Overexpression of full-length TCFs with Ras indicated that exogenous TCFs can regulate the CQE1-dependent transcription in a Ras-dependent manner. PD98059, an inhibitor of MAPK, significantly repressed the CQE1-dependent transcription. However, no serum response factor was detected by electrophoretic mobility shift assay using the CQE1 element. These results indicate that transcription of the Cctq gene is regulated by TCFs under the control of the Ras/MAPK pathway, probably independently of serum response factor.


Subject(s)
Chaperonins/genetics , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Serum Response Factor/pharmacology , Transcription Factors , Transcription, Genetic/physiology , Animals , Base Sequence , Binding Sites , Chaperonin Containing TCP-1 , Chaperonins/chemistry , Cytosol/physiology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Genes, Reporter , Genetic Complementation Test , Glycoproteins/metabolism , HeLa Cells , Humans , In Vitro Techniques , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Saposins , Sphingolipid Activator Proteins , Up-Regulation/genetics , ets-Domain Protein Elk-1 , ets-Domain Protein Elk-4 , ras Proteins/metabolism
12.
Blood ; 100(13): 4454-61, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12393577

ABSTRACT

Thrombin signaling in endothelial cells provides an important link between coagulation and inflammation. We report here that thrombin induces endogenous Egr-1 mRNA and Egr-1 promoter activity in primary human endothelial cells by approximately 6-fold and 3-fold, respectively. In transient transfection assays, deletion of the 3' cluster of serum response elements (SREs), but not the 5' cluster of SREs, resulted in a loss of thrombin response. When coupled to a heterologous core promoter, a region spanning the 3' SRE cluster contained information for thrombin response, whereas a region spanning the 5' SRE cluster had no such effect. A point mutation of the most proximal SRE (SRE-1), but not of the proximal Ets motif or upstream SREs, abrogated the response to thrombin. In electrophoretic mobility shift assays, nuclear extracts from thrombin-treated cells displayed increased binding of total and phosphorylated serum response factor (SRF) to SRE-1. Thrombin-mediated induction of Egr-1 was blocked by inhibitors of MEK1/2, but not by inhibitors of protein kinase C, phosphatidylinositol 3-kinase, or p38 mitogen-activated protein kinase (MAPK). Taken together, these data suggest that thrombin induces Egr-1 expression in endothelial cells by a MAPK-dependent mechanism that involves an interaction between SRF and SRE-1.


Subject(s)
DNA-Binding Proteins/genetics , Endothelium, Vascular/drug effects , Immediate-Early Proteins , Serum Response Element/drug effects , Serum Response Factor/pharmacology , Thrombin/pharmacology , Transcription Factors/genetics , Amino Acid Motifs , Cells, Cultured/cytology , Cells, Cultured/drug effects , Culture Media, Serum-Free/pharmacology , DNA-Binding Proteins/biosynthesis , Early Growth Response Protein 1 , Electrophoretic Mobility Shift Assay , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Genes, Reporter , Genes, Synthetic , Humans , Luciferases/biosynthesis , Luciferases/genetics , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Point Mutation , Protein Binding/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Transport/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/biosynthesis , Sequence Deletion , Transcription Factors/biosynthesis , Transcriptional Activation/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...