1
|
Arnoux F, Fina F, Lambert N, Balandraud N, Martin M, Ouafik L, Kanaan SB, Roudier J, Auger I. Newly Identified BRAF Mutation in Rheumatoid Arthritis. Arthritis Rheumatol 2017; 68:1377-83. [PMID: 26814611 DOI: 10.1002/art.39588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/07/2016] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA)-associated autoantibodies include those directed at the kinase site of BRAF (v-Raf murine sarcoma viral oncogene homolog B1), a serine-threonine kinase involved in the MAPK signaling pathway. To understand anti-BRAF immunization, we sought to identify BRAF mutations in the peripheral blood lymphocytes (PBLs) of patients with RA. METHODS We first cloned the major BRAF region known for mutations in the pCR2.1 vector, using genomic DNA from the PBLs of 8 RA patients. For each patient, 100 clones were sequenced. In 5 of 8 patients, we detected a new BRAF mutation in 1 clone. The frequency of this new mutation was evaluated by droplet digital polymerase chain reaction in PBLs from RA patients and controls. To test whether p.Val600Ala influences the kinase activity of BRAF, we developed an in vitro assay based on phosphorylation of MEK-1, a major BRAF substrate. RESULTS A BRAF mutation, p.Val600Ala, was identified in 1 of 8,000 PBLs and 1 of 6,000 T lymphocytes from RA patients and in 1 of 12,500 PBLs and 1 of 12,500 T lymphocytes from controls. The BRAF p.Val600Ala mutation was not correlated with the presence of anti-BRAF autoantibodies. The p.Val600Ala mutation activated phosphorylation of MEK-1 in vitro. CONCLUSION Most RA patients have a p.Val600Ala mutation in the BRAF gene. This mutation activates the kinase activity of BRAF. The p.Val600Ala mutation could activate the MAPK pathway, leading to the activation of T lymphocytes.
Collapse
Affiliation(s)
- Fanny Arnoux
- INSERM UMR-S 1097 and Aix Marseilles University, Marseilles, France
| | - Frederic Fina
- Assistance Publique-Hôpitaux de Marseilles, Marseilles, France
| | - Nathalie Lambert
- INSERM UMR-S 1097 and Aix Marseilles University, Marseilles, France
| | - Nathalie Balandraud
- INSERM UMR-S 1097, Aix Marseilles University, and Assistance Publique-Hôpitaux de Marseilles, Marseilles, France
| | - Marielle Martin
- INSERM UMR-S 1097 and Aix Marseilles University, Marseilles, France
| | | | - Sami B Kanaan
- INSERM UMR-S 1097 and Aix Marseilles University, Marseilles, France
| | - Jean Roudier
- INSERM UMR-S 1097, Aix Marseilles University, and Assistance Publique-Hôpitaux de Marseilles, Marseilles, France
| | - Isabelle Auger
- INSERM UMR-S 1097 and Aix Marseilles University, Marseilles, France
| |
Collapse
|
2
|
The lack of Raf-1 kinase feedback regulation enhances antiapoptosis in cancer cells. Oncogene 2016; 36:2014-2022. [PMID: 27841865 DOI: 10.1038/onc.2016.384] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 07/15/2016] [Accepted: 09/04/2016] [Indexed: 12/15/2022]
Abstract
Raf-1 has an important role in cellular antiapoptosis. So far, there is no solid evidence that shows that Raf-1 mutation is associated with cancer development. In the course of further study of Raf-1 signaling, we have reported that Raf-1 hyperphosphorylation inhibits its kinase activity toward its downstream mitogen-activated protein kinase kinase 1/2 (MEK1/2) and proposed a model for negative feedback regulation of Raf-1. Here, we show that there is no hyperphosphorylation in some cancer cells, which results in increased kinase activity and enhances the antiapoptotic ability. Inhibition of either Raf-1 or ALG-2 (apoptosis-linked gene 2) expression results in apoptosis signal-regulating kinase 1/c-Jun N-terminal kinase (ASK1/JNK) signaling activation, and cell sensitivity to chemotherapeutic reagents, indicating that inhibition of ASK1/JNK apoptotic signaling by Raf-1 is mediated by ALG-2. A previous report indicated that extracellular signal-regulated kinase 1/2 (ERK1/2) were responsible for Raf-1 hyperphosphorylation. However, our evidence shows that when ERK1/2 are activated and the Raf-1 gene is not mutated, Raf-1 is not hyperphosphorylated in these cells, indicating that ERK1/2 are not responsible for the Raf-1 hyperphosphorylation in these cancer cell lines. Surprisingly, we also found that Raf-1 is not a necessary kinase for MEK1/2 activation under normal tissue culture conditions, but is required for MEK1/2 activation under apoptosis-inducing conditions. Our research demonstrates that although Raf-1 gene is not mutated, an abnormality of Raf-1 kinase feedback regulation enhances its antiapoptotic function, and Raf-1 can still be a pharmaceutical target to increase chemotherapy or radiotherapy sensitivity in these cancer cells.
Collapse
|
3
|
Zhao GQ, Lin J, Hu LT, Yin XN, Wang Q, Xu Q, Li H. The role of Dectin-1/Raf-1 signal cascade in innate immune of human corneal epithelial cells against Aspergillus fumigatus infection. Int J Ophthalmol 2016; 9:1371-1375. [PMID: 27803850 DOI: 10.18240/ijo.2016.10.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022] Open
Abstract
AIM To investigate the expression of the v-raf-1 murine leukemia viral oncogene homolog 1 (Raf-1) and its role in the innate immune response of human corneal epithelial cells (HCECs) infected by Aspergillus fumigatus. METHODS HCECs were cultured in vitro. They were randomly divided into 4 groups, including control group, Aspergillus fumigatus group, GW5074 (an inhibitor of Raf-1) group and Laminarin [an inhibitor of Dendriti-cell-associated C-type lectin 1 (Dectin-1)] group. The protein expression level of total Raf-1 and p-Raf-1was measured by Western blot. The expression of IL-6 and IL-8 mRNA in each group was detected by real-time polymerase chain reaction. RESULTS In Aspergillus fumigatus group, total Raf-1 protein levels in HCECs remained unchanged at 5, 15, 30 and 45min after infection, while p-Raf-1 expression was significantly enhanced at 30min after infection compared with control group. However, the expression of p-Raf-1 was apparently declined after treated with GW5074 or Laminarin compared with Aspergillus fumigatus group. The expression levels of IL-6, IL-8 mRNA were significantly increased after stimulation with fumigatus compared with control group. Pre-treated with GW5074 significantly inhibited Aspergillus fumigatus-induced upregulation of IL-8 and IL-6. CONCLUSION Aspergillus fumigatus stimulation can elevate the expression of p-Raf-1 in HCECs in vitro. Dectin-1/Raf-1 signal pathway may play a role on regulating the expression of inflammatory cytokines, including IL-6 and IL-8.
Collapse
Affiliation(s)
- Gui-Qiu Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Li-Ting Hu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Xiao-Ni Yin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Qian Wang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Qiang Xu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Hui Li
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| |
Collapse
|
4
|
Auger I, Charpin C, Balandraud N, Martin M, Roudier J. Autoantibodies to PAD4 and BRAF in rheumatoid arthritis. Autoimmun Rev 2012; 11:801-3. [PMID: 22349616 DOI: 10.1016/j.autrev.2012.02.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes cartilage and bone destruction. The mechanisms leading to RA are unknown. There is currently no reliable cure for RA. Early treatment can reduce inflammation, joint damage and bone destruction. Thus, early diagnosis of RA is critical. However, definitive diagnosis of RA can be difficult. Immunologic tests that can be performed for the diagnosis of RA include detection of anti citrullinated protein antibodies (ACPAs). However, one third of RA patients have no ACPAs. To identify new autoantibodies in RA, we used the sera of RA patients to screen protein arrays containing 8000 human proteins. We found and validated two major autoantigens: PAD4 (peptidyl arginine deiminase 4) and BRAF (v raf murine sarcoma viral oncogene homolog B1) catalytic domain. We identified peptide targets of anti PAD4 and BRAF autoantibodies. We observed that anti PAD4 are inhibitory whereas anti BRAF stimulate BRAF activity. Anti PAD4 and anti BRAF antibodies may be used to diagnose RA, particularly in the absence of anti citrullinated protein antibodies.
Collapse
Affiliation(s)
- Isabelle Auger
- INSERM UMR 1097, Aix Marseille Université, Parc Scientifique de Luminy, Marseille, France
| | | | | | | | | |
Collapse
|
5
|
Charpin C, Martin M, Balandraud N, Roudier J, Auger I. Autoantibodies to BRAF, a new family of autoantibodies associated with rheumatoid arthritis. Arthritis Res Ther 2010; 12:R194. [PMID: 20955560 PMCID: PMC2991030 DOI: 10.1186/ar3165] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 09/08/2010] [Accepted: 10/18/2010] [Indexed: 01/08/2023] Open
Abstract
Introduction BRAF (v raf murine sarcoma viral oncogene homologue B1) is a serine-threonine kinase involved in the mitogen-activated protein kinase (MAPK) signalling pathway, known to be implicated in the production of pro-inflammatory cytokines. We have observed that sera from rheumatoid arthritis (RA) patients recognize the BRAF's catalytic domain, which encompasses amino acids 416 to 766. Here, we identify peptide targets of anti-BRAF autoantibodies and test whether anti-BRAF autoantibodies may interfere with BRAF kinase activity. Methods Anti-BRAF autoantibodies were detected by ELISA (enzyme-linked immunosorbent assay) in the serum of RA patients and controls, using 40 overlapping 20mer peptides encompassing the catalytic domain of BRAF as immunosorbents. To test whether autoantibodies to BRAF influence BRAF kinase activity, we developed an in vitro phosphorylation assay of MEK1 (mitogen extracellular regulated kinase), a major BRAF substrate. MEK1 phosphorylation by BRAF was tested in the presence of purified anti-BRAF autoantibodies from RA patients or control antibody. Results We found that one BRAF peptide, P25 (656 to 675), is specifically recognized by autoantibodies from RA patients. Of interest, anti-P25 autoantibodies are detected in 21% of anti-CCP (cyclic citrullinated peptides) negative RA patients. Anti-BRAF autoantibodies activate the in vitro phosphorylation of MEK1 mediated by BRAF. Conclusions Anti-BRAF autoantibodies from RA patients preferentially recognize one BRAF peptide: P25. Autoantibody responses to P25 are detected in 21% of anti-CCP negative RA patients. Most anti-BRAF autoantibodies activate BRAF kinase activity.
Collapse
Affiliation(s)
- Caroline Charpin
- INSERM UMR 639, Parc scientifique et technologique de Luminy, 163 avenue de Luminy, 13288 Marseille cedex 09, France.
| | | | | | | | | |
Collapse
|
6
|
Auger I, Balandraud N, Rak J, Lambert N, Martin M, Roudier J. New autoantigens in rheumatoid arthritis (RA): screening 8268 protein arrays with sera from patients with RA. Ann Rheum Dis 2009; 68:591-4. [PMID: 18957483 DOI: 10.1136/ard.2008.096917] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To identify new IgG autoantibodies in sera from patients with rheumatoid arthritis (RA). METHODS We tested serum samples from 19 patients with RA with given human leukocyte antigen (HLA)-DR genotypes, from 7 patients with spondylarthropathy, 2 patients with lupus, 4 patients with systemic sclerosis and 10 healthy individuals on 8268 human protein arrays. RESULTS We identified four antigens (peptidyl arginine deiminase 4 (PAD4), protein kinase Cbeta1 (PKCbeta1), phosphatylinositol 4 phosphate 5 kinase type II gamma (PIP4K2C) and v raf murine sarcoma viral oncogene homologue B1 catalytic domain (BRAF)) that were recognised almost uniquely by sera from patients with RA on protein arrays. Using purified proteins, we confirmed that PAD4 and BRAF are recognised almost uniquely by patients with RA. CONCLUSION We identified PAD4 and BRAF as RA specific autoantigens.
Collapse
Affiliation(s)
- I Auger
- INSERM UMR 639, Parc Scientifique de Luminy, APHM La Conception, Marseille, France.
| | | | | | | | | | | |
Collapse
|
7
|
Chaib H, Hoskins BE, Ashraf S, Goyal M, Wiggins RC, Hildebrandt F. Identification of BRAF as a new interactor of PLCε1, the protein mutated in nephrotic syndrome type 3. Am J Physiol Renal Physiol 2008; 294:F93-9. [DOI: 10.1152/ajprenal.00345.2007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Steroid-resistant nephrotic syndrome is a malfunction of the kidney glomerular filter that leads to proteinuria, hypoalbuminemia, edema, and renal failure. Recently, we identified recessive mutations in the phospholipase C epsilon 1 gene ( PLCE1) as a new cause of early-onset nephrotic syndrome and demonstrated interaction of PLCε1 with IQGAP1. To further elucidate the mechanism by which PLCE1 mutations cause nephrotic syndrome, we sought to identify new protein interaction partners of PLCε1. We utilized information from the genetic interaction network of C. elegans. It relates the PLCE1 ortholog ( plc-1) to the C. elegans ortholog ( lin-45) of human BRAF ( v-raf murine sarcoma viral oncogene homolog B1). We hypothesized that this may indicate a functional protein-protein interaction. Using GST pull down of HEK293T cell lysates in vitro and coimmunoprecipation of mouse kidney lysates in vivo, we show that BRAF interacts with PLCε1. By immunohistochemistry in rat kidney, we demonstrate that both proteins are coexpressed and colocalize in developing and mature glomerular podocytes, reporting for the first time the expression of BRAF in the glomerular podocyte.
Collapse
|
8
|
|
9
|
Nielsen C, Thastrup J, Bøttzauw T, Jäättelä M, Kallunki T. c-Jun NH2-terminal kinase 2 is required for Ras transformation independently of activator protein 1. Cancer Res 2007; 67:178-85. [PMID: 17210697 DOI: 10.1158/0008-5472.can-06-2801] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Active Ras oncogene is expressed in approximately 30% of human cancers. Yet, very little is known about the molecular mechanisms responsible for its transforming potential. Here, we show that H-Ras-mediated transformation requires isoform 2 of the c-Jun-NH(2)-terminal kinase (JNK). H-Ras-transduced JNK2-deficient (Jnk2-/-) murine embryonic fibroblasts (MEFs) were severely inhibited in colony formation and growth in soft agar in vitro as well as in tumor formation in immunodeficient mice as compared with corresponding Jnk1-/- and wild-type MEFs. Accordingly, the RNA interference-based depletion of JNK2 form wild-type MEFs also resulted in defective Ras transformation. The extra barrier against H-Ras transformation in Jnk2-/- MEFs was not due to their inability to inactivate p53 signaling because all JNK2-deficient MEF lines had lost p19(Arf). Furthermore, expression of the E6 protein of the human papilloma virus failed to overcome the transformation defect. It could, however, be overcome by coexpression of H-Ras with the SV40 large T antigen or c-Myc. Surprisingly, the H-Ras-transduced JNK2-deficient MEFs exhibited higher activity of activator protein-1 and higher levels of c-Jun expression compared with H-Ras-transduced JNK1-deficient or wild-type cells, indicating that the key target of JNK2 during Ras transformation was divergent from activator protein-1. These results clearly show that a single kinase, JNK2, could control Ras transformation and thus point out a vulnerable control point that may prove important for the tumor development in general.
Collapse
Affiliation(s)
- Christina Nielsen
- Apoptosis Department and Centre for Genotoxic Stress, Institute of Cancer Biology, Danish Cancer Society, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
10
|
Affiliation(s)
- R Schäfer
- Department of Pathology, University of Zurich, Switzerland
| |
Collapse
|
11
|
Autieri MV, Chen X. The ability of AIF-1 to activate human vascular smooth muscle cells is lost by mutations in the EF-hand calcium-binding region. Exp Cell Res 2005; 307:204-11. [PMID: 15922740 DOI: 10.1016/j.yexcr.2005.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2004] [Revised: 02/28/2005] [Accepted: 03/01/2005] [Indexed: 12/01/2022]
Abstract
Allograft Inflammatory Factor-1 (AIF-1) is a cytoplasmic calcium-binding protein expressed in vascular smooth muscle cells (VSMC) in response to injury or cytokine stimulation. AIF-1 contains a partially conserved EF-hand calcium-binding domain, and participates in VSMC activation by activation of Rac1 and induction of Granulocyte-Colony Stimulating Factor (G-CSF) expression; however, the mechanism whereby AIF-1 mediates these effects is presently uncharacterized. To determine if calcium binding plays a functional role in AIF-1 activity, a single site-specific mutation was made in the EF-hand calcium-binding domain to abrogate binding of calcium (AIF-1DeltaA), which was confirmed by calcium overlay. Functionally, similar to wild-type AIF-1, AIF-1DeltaA was able to polymerize F-actin in vitro. However, in contrast to wild-type AIF-1, over-expression of AIF-1DeltaA was unable to increase migration or proliferation of primary human VSMC. Further, it was unable to activate Rac1, or induce G-CSF expression to the degree as wild-type AIF-1. Taken together, modification of the wild-type EF-hand domain and native calcium-binding activity results in a loss of AIF-1 function. We conclude that appropriate calcium-binding potential is critical in AIF-1-mediated effects on VSMC pathophysiology, and that AIF-1 activity is mediated by Rac1 activation and G-CSF expression.
Collapse
Affiliation(s)
- Michael V Autieri
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Room 810, MRB 3420 N. Broad Street, Philadelphia PA 19140, USA.
| | | |
Collapse
|
12
|
Flaxenburg JA, Melter M, Lapchak PH, Briscoe DM, Pal S. The CD40-induced signaling pathway in endothelial cells resulting in the overexpression of vascular endothelial growth factor involves Ras and phosphatidylinositol 3-kinase. THE JOURNAL OF IMMUNOLOGY 2004; 172:7503-9. [PMID: 15187129 DOI: 10.4049/jimmunol.172.12.7503] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ligation of endothelial cell (EC) CD40 induces the expression of several proinflammatory cytokines as well as angiogenesis factors, including vascular endothelial growth factor (VEGF). Moreover, despite the reported importance of CD40 in cell-mediated immunity, little is known of the CD40-induced signaling pathways in EC. In this study, we have investigated the function of the Ras signaling pathway(s) for CD40-induced overexpression of VEGF. EC were transiently transfected with a full-length VEGF promoter-luciferase construct and a dominant-inhibitory mutant of Ras (Ras17N). Following transfection, ligation of CD40 with soluble CD40 ligand resulted in a significant increase in VEGF transcriptional activation, and the inhibitory mutant of Ras blocked this CD40-induced VEGF overexpression. Using EMSA and Western blot analysis, we demonstrated that CD40-dependent binding of nuclear protein(s) to the VEGF promoter and CD40-induced VEGF protein expression in EC were also inhibited by the Ras mutant. Immunoprecipitation studies revealed that ligation of CD40 on EC promoted an increased association of Ras with its effector molecules Raf, Rho, and phosphatidylinositol 3-kinase (PI3K). But, cotransfection of effector-loop mutants of Ras determined that only PI3K was functional for Ras-induced VEGF transcription. Also, wortmanin and a dominant-inhibitory mutant of PI3K inhibited CD40-induced overexpression of VEGF. Together these findings demonstrate that both Ras and PI3K are intermediaries in CD40-induced regulation of VEGF in EC. We believe our findings are of importance in several chronic inflammatory diseases, including atherosclerosis and allograft rejection associated with both CD40-CD40 ligand signaling as well as VEGF expression and function.
Collapse
Affiliation(s)
- Jesse A Flaxenburg
- Division of Nephrology, Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
13
|
Autieri MV. Inducible expression of the signal transduction protein 14-3-3gamma in injured arteries and stimulated human vascular smooth muscle cells. Exp Mol Pathol 2004; 76:99-107. [PMID: 15010287 DOI: 10.1016/j.yexmp.2003.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2003] [Indexed: 01/20/2023]
Abstract
Although the 14-3-3 family of proteins have been shown to be key signal transduction proteins involved in regulation of cellular growth and proliferation, little has been reported on their expression in pathophysiological states. We hypothesized that expression of one isoform, 14-3-3gamma, would also be increased in vascular proliferative diseases. We observed 14-3-3gamma expression induced in human coronary artery vasculopathy (CAV) as compared with coronary arteries isolated from normal and end-stage heart failure patients. 14-3-3gamma is acutely expressed in aortic medial smooth muscle cells in experimental models of arterial injury including rat cardiac allografts balloon angioplasty-injured swine coronary arteries. In each case, 14-3-3gamma protein expression is induced by 3 days and peaks at 7-10 days post-injury. Expression of this protein in cultured human vascular smooth muscle cells (VSMC) is associated with cytokine-induced VSMC activation, rather than direct injury to the VSMC themselves, and is unique among other 14-3-3 family proteins. Potential 14-3-3gamma protein-protein interactions are also differentially regulated by cytokine stimulation. This study indicates that 14-3-3gamma expression is induced in arterial trauma by cytokines, and suggests that this protein may play an important role in progression of vascular proliferative diseases.
Collapse
Affiliation(s)
- Michael V Autieri
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
14
|
Chen C, Dickman MB. Dominant active Rac and dominant negative Rac revert the dominant active Ras phenotype in Colletotrichum trifolii by distinct signalling pathways. Mol Microbiol 2004; 51:1493-507. [PMID: 14982641 DOI: 10.1111/j.1365-2958.2003.03932.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The small G-protein superfamily is an evolutionarily conserved group of GTPases that regulate diverse signalling pathways including pathways for growth and development in eukaryotes. Previously, we showed that dominant active mutation in the unique Ras gene (DARas) of the fungal phytopathogen Colletotrichum trifolii displays a nutrient-dependent phenotype affecting polarity, growth and differentiation. Signalling via the MAP kinase pathway is significantly impaired in this mutant as well. Here we describe the cloning and functional characterization of Rac (Ct-Rac1), a member of the Rho family of G proteins. Ct-Rac1 expression is downregulated by DARas under limiting nutrition. Co-expression of DARas with dominant active Rac (DARac) stimulates MAPK activation and restores the wild-type phenotype. Inhibition of MAPK activation suppresses phenotypic restoration suggesting Rac-mediated MAPK activation is responsible for reversion to the wild-type phenotype. We also examined the role of reactive oxygen species (ROS) in these genetic backgrounds. The DARas mutant strain generates high levels of ROS as determined by DCFH-DA fluorescence. Co-expression with DNRac decreases ROS generation to wild-type levels and restores normal fungal growth and development. Pretreatment of DARas with antioxidants or a cytosolic phospholipase A2 inhibitor also restores the wild-type phenotype. These findings suggest that Ras-mediated ROS generation is dependent on a Rac-cPLA(2)-linked signalling pathway. Taken together, this study provides evidence that Rac functions to restore the hyphal morphology of DARas by regulating MAPK activation and intracellular ROS generation.
Collapse
Affiliation(s)
- Changbin Chen
- Department of Plant Pathology, University of Nebraska-Lincoln, Nebraska 68583, USA
| | | |
Collapse
|
15
|
Ghosh S, Moore S, Bell RM, Dush M. Functional analysis of a phosphatidic acid binding domain in human Raf-1 kinase: mutations in the phosphatidate binding domain lead to tail and trunk abnormalities in developing zebrafish embryos. J Biol Chem 2003; 278:45690-6. [PMID: 12925535 DOI: 10.1074/jbc.m302933200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously, we and others identified a 35-amino acid segment within human Raf-1 kinase that preferentially binds phosphatidic acid. The presence of phosphatidic acid was found to be necessary for the translocation of Raf-1 to the plasma membrane. We have now employed a combination of alanine-scanning and deletion mutagenesis to identify the critical amino acid residues in Raf-1 necessary for interaction with phosphatidic acid. Progressive mutations within a tetrapeptide motif (residues 398-401 of human Raf-1) reduced and finally eliminated binding of Raf-1 to phosphatidic acid. We then injected zebrafish embryos with RNA encoding wild-type Raf-1 kinase or a mutant version with triple alanine mutations in the tetrapeptide motif and followed the morphological fate of embryonic development. Embryos with mutant but not wild-type Raf-1 exhibited defects in posterior axis formation exemplified by bent trunk and tail structures. Molecular evidence for lack of signaling through mutated Raf-1 was obtained by aberrant in situ hybridization of the ntl (no tail) gene, which functions downstream of Raf-1. Our results demonstrate that a functional phosphatidate binding site is necessary for Raf-1 function in embryonic development.
Collapse
Affiliation(s)
- Sujoy Ghosh
- GlaxoSmithKline, Genetics Research, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | |
Collapse
|
16
|
Pelegrinelli FF, Thirone AC, Gasparetti AL, Araujo EP, Velloso LA, Saad MJ. Early steps of insulin action in the skin of intact rats. J Invest Dermatol 2001; 117:971-6. [PMID: 11676840 DOI: 10.1046/j.0022-202x.2001.01473.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Insulin is an important regulator of growth and initiates its action by binding to its receptor, which undergoes tyrosyl autophosphorylation and further enhances its tyrosine kinase activity towards other intermediate molecules, including insulin receptor substrate 1, insulin receptor substrate 2, and Shc. Insulin receptor substrate proteins can dock various src-homology-2-domain-containing signaling proteins, such as the 85 kDa subunit of phosphatidylinositol 3 kinase and growth-factor-receptor-bound protein 2. The serine-threonine kinase is activated downstream to phosphatidylinositol 3 kinase. Shc protein has been shown to directly induce the association with growth-factor-receptor-bound protein 2 and downstream the activation of the mitogen-activated protein kinase. In this study we investigated insulin signal transduction pathways in skin of intact rats by immunoprecipitation and immunoblotting with specific antibodies, and also by immunohistochemistry with anti-insulin-receptor antibody. Our results showed that skin fragments clearly demonstrated the presence of insulin receptor in cell bodies of the epidermis and hair follicles and some faint staining was also detected in fibroblasts of the dermis. It was also observed that acute stimulation with insulin can induce tyrosyl phosphorylation of insulin receptor, that the insulin receptor substrates and Shc proteins serve as signaling molecules for insulin in skin of rats, and that insulin is able to induce association of insulin receptor substrate 1/phosphatidylinositol 3 kinase and Shc/growth-factor-receptor-bound protein 2 in this tissue, as well as phosphorylation of mitogen-activated protein kinase and serine-threonine kinase, demonstrating that proteins involved in early steps of insulin action are expressed in skin of intact rats and are quickly activated after insulin stimulation.
Collapse
Affiliation(s)
- F F Pelegrinelli
- Department of Internal Medicine, FCM, University of Campinas, Campinas, SP, Brazil
| | | | | | | | | | | |
Collapse
|
17
|
Moody TW, Leyton J, Chan D, Brenneman DC, Fridkin M, Gelber E, Levy A, Gozes I. VIP receptor antagonists and chemotherapeutic drugs inhibit the growth of breast cancer cells. Breast Cancer Res Treat 2001; 68:55-64. [PMID: 11678309 DOI: 10.1023/a:1017994722130] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The effects of vasoactive intestinal peptide (VIP) antagonists on breast cancer cells were investigated. (N-stearyl, norleucine17)VIP hybrid ((SN)VIPhyb) inhibited specific 125I-VIP binding to MCF7, SKBR3, T47D ZR75-1 and MDA-MB231 cells with high affinity (IC50 values of 0.03-0.06 microM). (SN)VIPhyb, 1 microM, inhibited the ability of 10 nM VIP to cause elevation of cAMP and to increase c-fos mRNA. Micromolar concentrations of (SN)VIPhyb inhibited the proliferation of MDA-MB231 or MCF7 cells using a MTT and clonogenic assay. Using a MTT assay, (SN)VIPhyb enhanced the ability of taxol and doxorubicin to inhibit breast cancer growth. Using nude mice bearing MDA-MB231 xenografts, VIPhyb potentiated the ability of taxol to inhibit proliferation. The results indicate that VIP receptor antagonists increase the ability of chemotherapeutic drugs to kill breast cancer cells.
Collapse
Affiliation(s)
- T W Moody
- Cell and Cancer Biology Department, Medicine Branch, National Cancer Institute, Rockville, MD 20850, USA.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Kwon EJ, Oh EJ, Kim YS, Hirose F, Ohno K, Nishida Y, Matsukage A, Yamaguchi M, Yoo MA. E2F-dependent transcription of the raf proto-oncogene during Drosophila development. Nucleic Acids Res 2001; 29:1808-14. [PMID: 11292854 PMCID: PMC31312 DOI: 10.1093/nar/29.8.1808] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
D-raf, a Drosophila homolog of the raf proto-oncogene, has diverse functions throughout development and is transcribed in a wide range of tissues, with high levels of expression in the ovary and in association with rapid proliferation. The expression pattern resembles those of S phase genes, which are regulated by E2F transcription factors. In the 5'-flanking region of D-raf, four sequences (E2F sites 1-4) similar to the E2F recognition sequence were found, one of them (E2F site 3) being recognized efficiently by Drosophila E2F (dE2F) in vitro. Transient luciferase expression assays confirmed activation of the D-raf gene promoter by dE2F/dDP. Expression of Draf-lacZ was greatly reduced in embryos homozygous for the dE2F mutation. These results suggest that dE2F is likely to be an important regulator of D-raf transcription.
Collapse
Affiliation(s)
- E J Kwon
- Department of Molecular Biology, College of Natural Science, Pusan National University, Pusan 609-735, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Pal S, Datta K, Khosravi-Far R, Mukhopadhyay D. Role of protein kinase Czeta in Ras-mediated transcriptional activation of vascular permeability factor/vascular endothelial growth factor expression. J Biol Chem 2001; 276:2395-403. [PMID: 11060301 DOI: 10.1074/jbc.m007818200] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF), a multifunctional cytokine, is regulated by different factors including degree of cell differentiation, hypoxia, and certain oncogenes namely, ras and src. The up-regulation of VPF/VEGF expression by Ras has been found to be through both transcription and mRNA stability. The present study investigates a novel pathway whereby Ras promotes the transcription of VPF/VEGF by activating protein kinase Czeta (PKCzeta). The Ras-mediated overexpression of VPF/VEGF was also found to be inhibited by using the antisense or the dominant-negative mutant of PKCzeta. In co-transfection assays, by overexpressing oncogenic Ha-Ras (12 V) and PKCzeta, there was an additive effect up to 4-fold in activation of Sp1-mediated VPF/VEGF transcription. It has been shown through electrophoretic mobility shift assay that Ras promoted the PKCzeta-induced binding of Sp1 to the VPF/VEGF promoter. In the presence of PDK-1, a major activating kinase for PKC, the Ras-mediated activation of VPF/VEGF promoter through PKCzeta was further increased, suggesting that PKCzeta can serve as an effector for both Ras and PDK-1. In other experiments, with the use of a dominant-negative mutant of phosphatidylinositol 3-kinase, the activation of VPF/VEGF promoter through Ras, PDK-1, and PKCzeta was completely repressed, indicating phosphatidylinositol 3-kinase as an important component of this pathway. Taken together, these data elucidate the signaling mechanism of Ras-mediated VPF/VEGF transcriptional activation through PKCzeta and also provide insight into PKCzeta and Sp1-dependent transcriptional regulation of VPF/VEGF.
Collapse
Affiliation(s)
- S Pal
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
20
|
Gupta S, Plattner R, Der CJ, Stanbridge EJ. Dissection of Ras-dependent signaling pathways controlling aggressive tumor growth of human fibrosarcoma cells: evidence for a potential novel pathway. Mol Cell Biol 2000; 20:9294-306. [PMID: 11094080 PMCID: PMC102186 DOI: 10.1128/mcb.20.24.9294-9306.2000] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Activation of multiple signaling pathways is required to trigger the full spectrum of in vitro and in vivo phenotypic traits associated with neoplastic transformation by oncogenic Ras. To determine which of these pathways are important for N-ras tumorigenesis in human cancer cells and also to investigate the possibility of cross talk among the pathways, we have utilized a human fibrosarcoma cell line (HT1080), which contains an endogenous mutated allele of the N-ras gene, and its derivative (MCH603c8), which lacks the mutant N-ras allele. We have stably transfected MCH603c8 and HT1080 cells with activating or dominant-negative mutant cDNAs, respectively, of various components of the Raf, Rac, and RhoA pathways. In previous studies with these cell lines we showed that loss of mutant Ras function results in dramatic changes in the in vitro phenotypic traits and conversion to a weakly tumorigenic phenotype in vivo. We report here that only overexpression of activated MEK contributed significantly to the conversion of MCH603c8 cells to an aggressive tumorigenic phenotype. Furthermore, we have demonstrated that blocking the constitutive activation of the Raf-MEK, Rac, or RhoA pathway alone is not sufficient to block the aggressive tumorigenic phenotype of HT1080, despite affecting a number of in vitro-transformed phenotypic traits. We have also demonstrated the possibility of bidirectional cross talk between the Raf-MEK-ERK pathway and the Rac-JNK or RhoA pathway. Finally, overexpression of activated MEK in MCH603c8 cells appears to result in the activation of an as-yet-unidentified target(s) that is critical for the aggressive tumorigenic phenotype.
Collapse
Affiliation(s)
- S Gupta
- Department of Microbiology and Molecular Genetics, College of Medicine, University of California-Irvine, Irvine, California 92697-4025, USA
| | | | | | | |
Collapse
|
21
|
Lu L, Ge Y, Li ZH, Dai MS, Broxmeyer HE. Enhancement of proliferation and differentiation of erythroid progenitors by co-transduction of erythropoietin receptor and H-ras cDNAS into single CD34(3+) cord blood cells. Bone Marrow Transplant 2000; 26:817-22. [PMID: 11081379 DOI: 10.1038/sj.bmt.1702620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Our previous studies have demonstrated that retrovirus-mediated gene transduction of either the human erythropoietin receptor (EpoR) or H-ras cDNA into single purified hematopoietic progenitor (HPC), CD34(3+), cells from cord blood (CB) resulted in increased numbers and sizes of erythroid cell containing colonies. We therefore evaluated if there were further effects when H-ras and EpoR genes were co-transduced into the same progenitor cells. Highly purified single sorted CD34(3+) CB cells were transduced with retroviral vectors encoding EpoR or H-ras cDNA. At the single cell level, and in response to stimulation by a combination of growth factors, including Epo, the number of colonies formed by BFU-E and CFU-GEMM was significantly increased in cells transduced with either single H-ras or EpoR cDNA compared to mock virus-transduced cells as previously described. Increased numbers of BFU-E, but not CFU-GEMM, colonies were produced from cells simultaneously co-transduced with both EpoR and Hras genes. Little or no growth was seen in transduced cells without exogenously added cytokines. The size of all types of colonies including CFU-GM was increased in cells transduced with H-ras and/or EpoR cDNAs, and the greatest increase was noticed in cells co-transduced with both genes. Integration and expression of either gene in individual colonies as assessed by PCR and RT-PCR analysis were 45-62% and 48-58%, respectively, with approximately 31% of the cells containing and expressing both genes. These results add to information suggesting an enhancing interacting role of H-ras and EpoR in erythroid proliferation/differentiation.
Collapse
Affiliation(s)
- L Lu
- Department of Microbiology/lmmunology, Indiana University School of Medicine, Indianapolis 46202-5254, USA
| | | | | | | | | |
Collapse
|
22
|
Martínez-Lorenzo MJ, Anel A, Monleón I, Sierra JJ, Piñeiro A, Naval J, Alava MA. Tyrosine phosphorylation of the p85 subunit of phosphatidylinositol 3-kinase correlates with high proliferation rates in sublines derived from the Jurkat leukemia. Int J Biochem Cell Biol 2000; 32:435-45. [PMID: 10762069 DOI: 10.1016/s1357-2725(99)00142-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A prominent tyrosine phosphorylated protein of 85 kDa (p85) was detected in highly proliferative sublines derived from the Jurkat T cell leukemia. We undertook a study to characterize the identity of this protein and its possible role in the hyperproliferative phenotypes observed. Using immunoblot and immunoprecipitation techniques, this protein was characterized as the p85 regulatory subunit of phosphatidylinositol 3-kinase. Cell proliferation and p85 tyrosine phosphorylation was not affected by tyrphostin AG-490, an inhibitor of Jak kinases, wortmannin or LY294002, inhibitors of the activity of the catalytic phosphatidylinositol 3-kinase subunit. Herbimycin-A and PPI, inhibitors of src-like protein tyrosine kinases, and genistein, a general tyrosine kinase inhibitor, inhibited p85 tyrosine phosphorylation and induced cell death in the sublines. PD98059, an inhibitor of Mek, inhibited cell growth of the sublines, but not that of the parental cells. It was concluded that tyrosine phosphorylation of p85 is associated with highly proliferative tumoral phenotypes, at least in T cell leukemias, independent of the phosphatidylinositol 3-kinase activity of the catalytic subunit.
Collapse
Affiliation(s)
- M J Martínez-Lorenzo
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Spain
| | | | | | | | | | | | | |
Collapse
|
23
|
Differential roles of Ca(2+)/calmodulin-dependent protein kinase II and mitogen-activated protein kinase activation in hippocampal long-term potentiation. J Neurosci 1999. [PMID: 10493730 DOI: 10.1523/jneurosci.19-19-08292.1999] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The roles of Ca(2+)/calmodulin-dependent protein kinase II (CaM kinase II) and mitogen-activated protein kinase (MAPK) in long-term potentiation (LTP) were investigated in the CA1 area of hippocampal slices, using electrophysiological and biochemical approaches. A brief high-frequency stimulation, but not low-frequency stimulation, delivered to Schaffer collateral/commissural afferents produced a stable LTP and activated both CaM kinase II and 42 kDa MAPK. Different from the activity of CaM kinase II, the increase in MAPK activity was transient. At a concentration of 50 microM, but not of 30 microM, PD098059, a potent inhibitor of MAPK kinase, markedly inhibited the induction of LTP. Although the two concentrations had similar inhibitory effects on MAPK activity, only 50 microM PD098059 suppressed the activation of CaM kinase II. Application of calmidazolium, an antagonist of calmodulin, blocked both CaM kinase II activation and the LTP induction without affecting the increase in 42 kDa MAPK activity. Application of neurotrophin brain-derived neurotrophic factor (BDNF) promoted the induction of LTP, with concomitant activation of CaM kinase II. Under the same conditions, BDNF failed to activate MAPK in hippocampal slices. These results indicate that, although the LTP induction is accompanied by increases in two kinase activities, only CaM kinase II activation is required for this event.
Collapse
|
24
|
Bland KI. The 1999 James Ewing Lecture: in pursuit of molecules of oncogenesis and neoplastic therapy. Ann Surg Oncol 1999; 6:528-41. [PMID: 10493620 DOI: 10.1007/s10434-999-0528-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Breast cancer is a progressive, phenotypic, genetic and epigenetic alteration, induced by various stimulants of which the principal steroid hormone, estrogen, initiates and promotes neoplastic transformation of normal ductal mammary epithelium to acquire distinct biochemical and molecular characteristics. The molecular events that characterize oncogenesis with dysregulated physiologic states represent an area of intense translational research. The progress of the last century suggests that the targeted responses initiated by this steroid hormone can be attenuated and modified with anti-estrogen therapies or through applied intervention with selective estrogen receptor modulation. New pharmacological, genetic, and biological agents will undoubtedly afford clinical investigators of the next millennium the opportunity to provide major impact on the disease progression of this neoplasm, through these targeted endocrine responses for estrogen.
Collapse
Affiliation(s)
- K I Bland
- Department of Surgery, Brown University School of Medicine, Providence, Rhode Island, USA
| |
Collapse
|
25
|
Improta-Brears T, Ghosh S, Bell RM. Mutational analysis of Raf-1 cysteine rich domain: requirement for a cluster of basic aminoacids for interaction with phosphatidylserine. Mol Cell Biochem 1999; 198:171-8. [PMID: 10497893 DOI: 10.1023/a:1006981411691] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Activation of Raf-1 kinase is preceded by a translocation of Raf-1 to the plasma membrane in response to external stimuli. The membrane localization of Raf-1 is facilitated through its interaction with activated Ras and with membrane phospholipids. Previous evidence suggests that the interaction of Raf-1 with Ras is mediated by two distinct domains within the N-terminal region of Raf-1 comprising amino acid residues 51-131 and residues 139-184, the latter of which codes for a zinc containing cysteine-rich domain. The cysteine-rich domain of Raf-1 is also reported to associate with other proteins, such as 14-3-3, and for selectively binding acidic phospholipids, particularly phosphatidylserine (PS). In the present study, we have investigated the consequences of progressive deletions and point mutations within the cysteine-rich domain of Raf-1 on its ability to bind PS. A reduced interaction with PS was observed in vitro for all deletion mutants of Raf-1 expressed either as full-length proteins or as fragments containing the isolated cysteine-rich domain. In particular, the cluster of basic amino acids R143, K144, and K148 appeared to be critical for interaction with PS, since substitution of all three residues to alanine resulted in a protein that failed to interact with liposomes enriched for PS. Expression of Raf-1 in vivo, containing point mutations in the cysteine-rich domain resulted in a truncated polypeptide that lacked both the Ras and PS binding sites and could no longer translocate to the plasma membrane upon serum stimulation. These results indicate that the basic residues 143, 144 and 148 in the anterior half of Raf-1 cysteine-rich domain play a role in the association with the lipid bilayer and possibly in protein stability, therefore they might contribute to Raf-1 localization and subsequent activation.
Collapse
Affiliation(s)
- T Improta-Brears
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
26
|
Xia K, Lee RS, Narsimhan RP, Mukhopadhyay NK, Neel BG, Roberts TM. Tyrosine phosphorylation of the proto-oncoprotein Raf-1 is regulated by Raf-1 itself and the phosphatase Cdc25A. Mol Cell Biol 1999; 19:4819-24. [PMID: 10373531 PMCID: PMC84280 DOI: 10.1128/mcb.19.7.4819] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There is a growing body of evidence demonstrating that Raf-1 is phosphorylated on tyrosines upon stimulation of a variety of receptors. Although detection of Raf-1 tyrosine phosphorylation has remained elusive, genetic analyses have demonstrated it to be important for Raf-1 activation. Here we report new findings which indicate that Raf-1 tyrosine phosphorylation is regulated in vivo. In both a mammalian and baculovirus expression system, a kinase-inactive allele of Raf-1 was found to be tyrosine phosphorylated at levels much greater than that of wild-type Raf-1. The level of tyrosine phosphate on Raf-1 was markedly increased upon treatment with phosphatase inhibitors either before or after cell lysis. Cdc25A was found to dephosphorylate Raf-1 on tyrosines that resulted in a significant decrease in Raf-1 kinase activity. In NIH 3T3 cells, coexpression of wild-type Raf-1 and phosphatase-inactive Cdc25A led to a marked increase in Raf-1 tyrosine phosphorylation in response to platelet-derived growth factor. These data suggest that the tyrosine phosphorylation of Raf-1 is regulated not only by itself but also by Cdc25A.
Collapse
Affiliation(s)
- K Xia
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
27
|
Autieri MV, Carbone CJ. 14-3-3Gamma interacts with and is phosphorylated by multiple protein kinase C isoforms in PDGF-stimulated human vascular smooth muscle cells. DNA Cell Biol 1999; 18:555-64. [PMID: 10433554 DOI: 10.1089/104454999315105] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It has recently been demonstrated that some members of the 14-3-3 protein family play an important role in signal transduction leading to cellular proliferation. We have previously shown that expression of 14-3-3gamma is induced by growth factors in human vascular smooth muscle cells (VSMC). In this study, we cloned the human homolog of 14-3-3gamma and observed many potential phosphorylation sites, suggesting the potential for post-translational modification. In VSMC treated with platelet-derived growth factor (PDGF), 14-3-3gamma protein was expressed and phosphorylated in an activation-dependent manner. Platelet-derived growth factor-induced phosphorylation could be inhibited by phosphokinase C (PKC) inhibitory compounds, and 14-3-3gamma could be phosphorylated in the absence of PDGF by compounds that activate PKC. We also demonstrated interaction between 14-3-3gamma and several PKC isoforms (alpha, beta, gamma, theta, and delta), implicating these PKC family isoforms as the kinases responsible for PDGF-induced 14-3-3gamma phosphorylation. We found that 14-3-3gamma interacted with the signal transduction protein Raf-1, suggesting that 14-3-3gamma provides a link between this protein and PKC. Thus, 14-3-3gamma may represent a signal transduction protein that is regulated transcriptionally and post-transcriptionally by growth factors.
Collapse
Affiliation(s)
- M V Autieri
- Department of Cardiology and Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA.
| | | |
Collapse
|
28
|
Abstract
VIP1 receptors are present in breast cancer cells. VIP elevates the cAMP and stimulates nuclear oncogene expression in MCF-7 cells. VIPhybrid is a VIP receptor antagonist that inhibits breast cancer proliferation. A VIP analog has been developed for imaging breast tumors. Therefore VIP1 receptors may be utilized for the early detection and treatment of breast cancer.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/pathology
- Breast Neoplasms/physiopathology
- Cyclic AMP/metabolism
- Female
- Humans
- Mice
- Mice, Nude
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Pituitary Hormone/analysis
- Receptors, Pituitary Hormone/physiology
- Receptors, Vasoactive Intestinal Peptide/analysis
- Receptors, Vasoactive Intestinal Peptide/physiology
- Receptors, Vasoactive Intestinal Peptide, Type II
- Receptors, Vasoactive Intestinal Polypeptide, Type I
- Tissue Distribution
- Transplantation, Heterologous
- Tumor Cells, Cultured
- Vasoactive Intestinal Peptide/pharmacokinetics
- Vasoactive Intestinal Peptide/physiology
Collapse
Affiliation(s)
- T W Moody
- Medicine Branch, National Cancer Institute, Rockville, Maryland, USA
| | | | | | | | | |
Collapse
|
29
|
Pratt MA, Satkunaratnam A, Novosad DM. Estrogen activates raf-1 kinase and induces expression of Egr-1 in MCF-7 breast cancer cells. Mol Cell Biochem 1998; 189:119-25. [PMID: 9879662 DOI: 10.1023/a:1006827015320] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We have investigated whether the raf-1 kinase, a downstream mediator of both receptor tyrosine kinase and protein kinase C signalling, is activated by estrogen (E2) in an estrogen receptor positive human breast cancer cell line. Autophosphorylation of raf-1 kinase was studied after treatment of MCF-7 cells with E2. E2-deprived cells contained low levels of raf-1 kinase activity. Treatment of cells for 1 min with E2 resulted in raf-1 autophosphorylation which was maximal within 5 min. Western blot analysis showed that raf-1 undergoes an electrophoretic mobility shift following E2 treatment. Egr-1 is a zinc finger-containing transcription factor which is expressed in association with raf-1 activation. Untreated MCF-7 cells expressed low levels of Egr-1 while E2 treatment resulted in an induction of egr-1 mRNA expression. These kinetics followed closely behind the E2 induction of c-myc mRNA. Egr-1 protein was similarly low in E2-deprived MCF-7 cells and was transiently increased following E2 treatment. Several studies have suggested that kinase activity may play a role in estrogen receptor (ER) activation. While activated v-raf failed to augment ER activation of transcription in transient transfection assays, a dominant negative mutant of raf-1 inhibited E2-induced transcription by 50% primarily as a result of increased baseline levels of E2 independent transcription. The results show that E2 can induce raf-1 kinase activity in MCF-7 breast cancer cells associated with the expression of an early growth response gene and modulation of ER signalling.
Collapse
Affiliation(s)
- M A Pratt
- Department of Pharmacology, University of Ottawa, Ontario, Canada
| | | | | |
Collapse
|
30
|
Atkins CM, Selcher JC, Petraitis JJ, Trzaskos JM, Sweatt JD. The MAPK cascade is required for mammalian associative learning. Nat Neurosci 1998; 1:602-9. [PMID: 10196568 DOI: 10.1038/2836] [Citation(s) in RCA: 874] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitogen-activated protein kinase (MAPK) is an integral component of cellular signaling during mitogenesis and differentiation of mitotic cells. Recently MAPK activation in post-mitotic cells has been implicated in hippocampal long-term potentiation (LTP), a potential cellular mechanism of learning and memory. Here we investigate the involvement of MAPK in learning and memory in behaving animals. MAPK activation increased in the rat hippocampus after an associative learning task, contextual fear conditioning. Two other protein kinases known to be activated during hippocampal LTP, protein kinase C and alpha-calcium/calmodulin protein kinase II, also were activated in the hippocampus after learning. Inhibition of the specific upstream activator of MAPK, MAPK kinase (MEK), blocked fear conditioning. Thus, classical conditioning in mammals activates MAPK, which is necessary for consolidation of the resultant learning.
Collapse
Affiliation(s)
- C M Atkins
- Division of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
31
|
de Boland AR, Norman AW. 1alpha,25(OH)2-vitamin D3 signaling in chick enterocytes: enhancement of tyrosine phosphorylation and rapid stimulation of mitogen-activated protein (MAP) kinase. J Cell Biochem 1998; 69:470-82. [PMID: 9620173 DOI: 10.1002/(sici)1097-4644(19980615)69:4<470::aid-jcb8>3.0.co;2-k] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The steroid hormone 1alpha,25(OH)2-vitamin D3 (1alpha,25(OH)2D3) generates biological responses in intestinal and other cells via both genomic and rapid, nongenomic signal transduction pathways. We examined the hypothesis that 1alpha,25(OH)2D3 action in chick enterocytes may be linked to pathways involving tyrosine phosphorylation. Brief exposure of isolated chick enterocytes to 1alpha,25(OH)2D3 demonstrated increased tyrosine phosphorylation of several cellular proteins (antiphosphotyrosine immunoblots of whole cell lysates) with prominent bands at 42-44, 55-60, and 105-120 Kda. The 42-44 Kda bands comigrated with mitogen-activated protein (MAP) kinase (immunoblotting with anti-MAP kinase antibody) The response occurred within 30 s, peaked at 1 min, and was dose-dependent (0.01-10 nM), with maximal stimulation at 1 nM (three- to fivefold). This effect was specific for 1alpha,25(OH)2D3 since its metabolic precursors 25(OH)D3 and vitamin D3 did not increase MAP kinase tyrosine phosphorylation. The tyrosine kinase inhibitor, genistein, blocked 1alpha,25(OH)2D3-induced tyrosine phosphorylation of MAP kinase, while staurosporine, a PKC inhibitor, attenuated the hormone's effects by 30%. We have evaluated the ability of 1alpha,25(OH)2D3 analogs, which have complete flexibility around the 6,7 carbon-carbon bond (6F) or which are locked in either the 6-s-cis (6C) or the 6-s-trans (6T) shape(s), to activate MAP kinase. Thus, two 6F and one 6C analog stimulated while one 6T analog did not stimulate MAP kinase tyrosine phosphorylation. In addition, 1beta,25(OH)2D3, a known antagonist of 1alpha,25(OH)2D3-mediated rapid responses, blocked the hormone effects on MAP kinase. We conclude that 1alpha,25(OH)2D3 and analogs which can achieve the 6-s-cis shape (6F and 6C) can increase tyrosine phosphorylation and activation of MAP kinase in chick enterocytes.
Collapse
Affiliation(s)
- A R de Boland
- Departamento de Biologia, Bioquimica, y Farmacia, Universidad Nacional del Sur, Bahia Blanca, Argentina
| | | |
Collapse
|
32
|
Briant L, Robert-Hebmann V, Sivan V, Brunet A, Pouysségur J, Devaux C. Involvement of Extracellular Signal-Regulated Kinase Module in HIV-Mediated CD4 Signals Controlling Activation of Nuclear Factor-κB and AP-1 Transcription Factors. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.4.1875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
Although the molecular mechanisms by which the HIV-1 triggers either T cell activation, anergy, or apoptosis remain poorly understood, it is well established that the interaction of HIV-1 envelope glycoproteins with cell surface CD4 delivers signals to the target cell, resulting in activation of transcription factors such as NF-κB and AP-1. In this study, we report the first evidence indicating that kinases MEK-1 (MAP kinase/Erk kinase) and ERK-1 (extracellular signal-regulated kinase) act as intermediates in the cascade of events that regulate NF-κB and AP-1 activation upon HIV-1 binding to cell surface CD4. We found that CEM cells transfected with dominant negative forms of MEK-1 or ERK-1 do not display NF-κB activation after HIV-1 binding to CD4. In contrast, NF-κB activation was observed in these cells after PMA stimulation. Although the different cell lines studied expressed similar amounts of CD4 and p56lck, HIV-1 replication and HIV-1-induced apoptosis were slightly delayed in cells expressing dominant negative forms of MEK-1 or ERK-1 compared with parental CEM cells and cells expressing a constitutively active mutant form of MEK-1 or wild-type ERK-1. In light of recently published data, we propose that a positive signal initiated following oligomerization of CD4 by the virus is likely to involve a recruitment of active forms of p56lck, Raf-1, MEK-1, and ERK-1, before AP-1 and NF-κB activation.
Collapse
Affiliation(s)
- Laurence Briant
- *Centre de Recherches de Biochimie Macromoléculaire, Laboratoire d’Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France; and
| | - Véronique Robert-Hebmann
- *Centre de Recherches de Biochimie Macromoléculaire, Laboratoire d’Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France; and
| | - Virginie Sivan
- *Centre de Recherches de Biochimie Macromoléculaire, Laboratoire d’Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France; and
| | | | | | - Christian Devaux
- *Centre de Recherches de Biochimie Macromoléculaire, Laboratoire d’Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France; and
| |
Collapse
|
33
|
Indolfi C, Avvedimento EV, Rapacciuolo A, Esposito G, Di Lorenzo E, Leccia A, Pisani A, Chieffo A, Coppola A, Chiariello M. In vivo gene transfer: prevention of neointima formation by inhibition of mitogen-activated protein kinase kinase. Basic Res Cardiol 1997; 92:378-84. [PMID: 9464861 DOI: 10.1007/bf00796211] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The mitogen-activated protein kinase kinase (MAPKK) is a protein downstream ras which is rapidly activated in cells stimulated with various extracellular signals. These proteins are believed to play a pivotal role in integrating and transmitting transmembrane signals required for cell growth. METHODS AND RESULTS To study the effect of inhibition of MAPKK on smooth muscle cell (SMC) proliferation in vivo after vascular injury, we performed experimental balloon angioplasty using the standard Clowes technique in male Wistar rats 14-weeks old. The animals did not receive any treatment after vascular injury (N = 6) or were randomly assigned to receive, after balloon injury, a 30% (w/v) pluronic gel solution applied to the injured carotid artery, containing respectively: 1) no plasmid DNA (n = 10); 2) RSV-lacZ (encoding the beta-galactosidase gene) as control gene without effects on SMC proliferation (n = 10); 3) Tg-CAT (encoding cloramphenicol acetyl-transferase gene under the control of thyreoglobulin promoter) as an additional control gene without effects on SMC proliferation (n = 7): 4) a negative mutant of Mitogen-Activated Protein Kinase Kinase (MAPKK-) (n = 13). Fourteen days after vascular injury, carotid arteries were removed and cross sections were cut and stained with hematoxylin/eosin. Morphometric analysis demonstrated, in the MAPKK- treated rats, a significant reduction of both neointima (0.096+/-.018 mm2 vs. 0.184+/-0.019 mm2, p < 0.01) and neointima/media ratio (0.603+/-0.103 vs. 1.471+/-0.161, p < 0.01) compared to control DNA. CONCLUSIONS The inhibition of MAPKK, by a dominant inhibitor mutant gene, prevents the SMC proliferation after vascular injury in vivo.
Collapse
Affiliation(s)
- C Indolfi
- Division of Cardiology, Federico II University, Napoli, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The delineation of gene function has always been an intensive subject of investigations. Recent advances in the synthesis and chemistry of oligonucleotides have now made these molecules important tools to study and identify gene function and regulation. Modulation of gene expression using oligonucleotides has been targeted at different levels of the cellular machinery. Triplex forming oligonucleotides, as well as peptide nucleic acids, have been used to inhibit gene expression at the level of transcription; after binding of these specific oligonucleotides, conformational change of the DNA's helical structure prevents any further DNA/protein interactions necessary for efficient transcription. Gene regulation can also be achieved by targeting the translation of mRNAs. Antisense oligonucleotides have been used to down-regulate mRNA expression by annealing to specific and determined region of an mRNA, thus inhibiting its translation by the cellular machinery. The exact mechanism of this type of inhibition is still under intense investigation and is thought to be related to the activation of RNase H, a ribonuclease that is widely available that can cleave the RNA/DNA duplex, thus making it inactive. Another well-characterized means of interfering with the translation of mRNAs is the use of ribozymes. Ribozymes are small catalytic RNAs that possess both site specificity and cleavage capability for an mRNA substrate, inhibiting any further protein formation. This review describes how these different oligonucleotides can be used to define gene function and discusses in detail their chemical structure, mechanism of action, advantages and disadvantages, and their applications.
Collapse
Affiliation(s)
- L D Curcio
- Department of General and Oncologic Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | | |
Collapse
|
35
|
Zhong C, Kinch MS, Burridge K. Rho-stimulated contractility contributes to the fibroblastic phenotype of Ras-transformed epithelial cells. Mol Biol Cell 1997; 8:2329-44. [PMID: 9362072 PMCID: PMC25711 DOI: 10.1091/mbc.8.11.2329] [Citation(s) in RCA: 133] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/1997] [Accepted: 08/25/1997] [Indexed: 02/05/2023] Open
Abstract
Oncogenic transformation of cells alters their morphology, cytoskeletal organization, and adhesive interactions. When the mammary epithelial cell line MCF10A is transformed by activated H-Ras, the cells display a mesenchymal/fibroblastic morphology with decreased cell-cell junctions but increased focal adhesions and stress fibers. We have investigated whether the transformed phenotype is due to Rho activation. The Ras-transformed MCF10A cells have elevated levels of myosin light chain phosphorylation and are more contractile than their normal counterparts, consistent with the activation of Rho. Furthermore, inhibitors of contractility restore a more normal epithelial phenotype to the Ras-transformed MCF10A cells. However, inhibiting Rho by microinjection of C3 exotransferase or dominant negative RhoA only partially restores the normal phenotype, in that it fails to restore normal junctional organization. This result prompted us to examine the effect that inhibiting Rho would have on the junctions of normal MCF10A cells. We have found that inhibiting Rho by C3 microinjection leads to a disruption of E-cadherin cytoskeletal links in adherens junctions and blocks the assembly of new adherens junctions. The introduction of constitutively active Rho into normal MCF10A cells did not mimic the Ras-transformed phenotype. Thus, these results lead us to conclude that some, but not all, characteristics of Ras-transformed epithelial cells are due to activated Rho. Whereas Rho is needed for the assembly of adherens junctions, high levels of activated Rho in Ras-transformed cells contribute to their altered cytoskeletal organization. However, additional events triggered by Ras must also be required for the disruption of adherens junctions and the full development of the transformed epithelial phenotype.
Collapse
Affiliation(s)
- C Zhong
- Department of Cell Biology and Anatomy, University of North Carolina at Chapel Hill 27599-7090, USA
| | | | | |
Collapse
|
36
|
Font de Mora J, Porras A, Ahn N, Santos E. Mitogen-activated protein kinase activation is not necessary for, but antagonizes, 3T3-L1 adipocytic differentiation. Mol Cell Biol 1997; 17:6068-75. [PMID: 9315666 PMCID: PMC232456 DOI: 10.1128/mcb.17.10.6068] [Citation(s) in RCA: 140] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In 3T3-L1 fibroblasts, Ras proteins mediate both insulin-induced differentiation to adipocytes and its activation of cytosolic serine/threonine kinases, including Raf-1 kinase, mitogen-activated protein kinase (MAPK), and Rsk. Here, we report that insulin- and Ras-induced activation of MAPK is not required for the differentiation process and in fact antagonizes it. The treatment of 3T3-L1 preadipocytes with MEK-specific inhibitor PD98059 blocked insulin- and Ras-induced MAPK activation but had no effect on or slightly enhanced adipocytic differentiation. Tumor necrosis factor alpha (TNF-alpha), an inhibitor of insulin-stimulated adipogenesis, activated MAPK in 3T3-L1 cells. PD98059 treatment blocked MAPK activation by TNF-alpha and reversed the blockade of adipogenesis mediated by low (1 ng/ml) TNF-alpha concentrations. 3T3-L1 transfectants containing hyperactivated MEK1 or overexpressed MAPK displayed impaired adipocytic differentiation. PD98059 treatment also reversed the blockade of differentiation in MEK1 transfectants. These results indicate that MAPK does not promote but can contribute to inhibition of the process of adipocytic differentiation of 3T3-L1 cells.
Collapse
Affiliation(s)
- J Font de Mora
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
37
|
Roux P, Gauthier-Rouvière C, Doucet-Brutin S, Fort P. The small GTPases Cdc42Hs, Rac1 and RhoG delineate Raf-independent pathways that cooperate to transform NIH3T3 cells. Curr Biol 1997; 7:629-37. [PMID: 9285711 DOI: 10.1016/s0960-9822(06)00289-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ras-mediated transformation of mammalian cells has been shown to activate multiple signalling pathways, including those involving mitogen-activated protein kinases and the small GTPase Rho. Members of the Rho family affect cell morphology by controlling the formation of actin-dependent structures: specifically, filopodia are induced by Cdc42Hs, lamellipodia and ruffles by Rac, and stress fibers by RhoA. In addition, Rho GTPases are involved in progression through the G1 phase of the cell cycle, and Rac1 and RhoA have recently been directly implicated in the morphogenic and mitogenic responses to transformation by oncogenic Ras. In order to examine the cross-talk between Ras and Rho proteins, we investigated the effects on focus-forming activity and cell growth of the Rho-family members Cdc42Hs, Rac1 and RhoG by expressing constitutively active or dominant-negative forms in NIH3T3 cells. RESULTS Expression of Rac1 or RhoG modulated the saturation density to which the cells grew, probably by affecting the level of contact inhibition. Although all three GTPases were required for cell transformation mediated by Ras but not by constitutively active Raf, the selective activation of each GTPase was not sufficient to induce the formation of foci. The coordinated activation of Cdc42Hs, RhoG and Rac1, however, elicited a high focus-forming activity, independent of the mitogen-activated ERK and JNK protein kinase pathways. CONCLUSIONS Ras-mediated transformation induces extensive changes in cell morphology which require the activity of members of the Rho family of GTPases. Our data show that the pattern of coordinated Rho family activation that elicits a focus-forming activity in NIH3T3 cells is distinct from the regulatory cascade that has been proposed for the control of actin-dependent structures in Swiss 3T3 cells.
Collapse
Affiliation(s)
- P Roux
- Institut de Génétique Moléculaire, C.N.R.S. UMR5535, 1919 route de Mende, F-34293 Montpellier cedex 5, France.
| | | | | | | |
Collapse
|
38
|
Abstract
BACKGROUND KSR (kinase suppressor of Ras) is a recently identified putative protein kinase that positively mediates the Ras signaling pathway in the invertebrates Caenorhabditis elegans and Drosophila melanogaster. The function of vertebrate KSR is not well characterized biochemically or biologically. RESULTS We examined the physiological role of KSR in vertebrate signal transduction using Xenopus laevis oocytes. Overexpression of KSR, in combination with overexpression of the intracellular dimeric protein 14-3-3, induced Xenopus oocyte meiotic maturation and cdc2 kinase activation; the effect of KSR and 14-3-3 on oocyte maturation was blocked by co-expression of dominant-negative Raf-1. We noted that KSR contains multiple potential binding sites for 14-3-3, and we used the yeast two-hybrid system and co-immunoprecipitation experiments to show that KSR can bind to 14-3-3. Furthermore, we demonstrated that KSR can form a complex with Raf kinase both in vitro and in cultured cells. Cell fractionation studies revealed that KSR formed a complex with 14-3-3 in both the membrane and cytoplasmic fractions of cell lysates; however, KSR only formed a complex with Raf-1 in the membrane fraction. CONCLUSIONS Our finding suggest that KSR, 14-3-3 and Raf form an oligomeric signaling complex and that KSR positively regulates the Ras signaling pathway in vertebrate organisms.
Collapse
Affiliation(s)
- H Xing
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
39
|
Ryu JR, Choi TY, Kwon EJ, Lee WH, Nishida Y, Hayashi Y, Matsukage A, Yamaguchi M, Yoo MA. Transcriptional regulation of the Drosophila-raf proto-oncogene by the DNA replication-related element (DRE)/DRE-binding factor (DREF) system. Nucleic Acids Res 1997; 25:794-9. [PMID: 9016631 PMCID: PMC146497 DOI: 10.1093/nar/25.4.794] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The DRE/DREF system plays an important role in transcription of DNA replication genes such as those encoding the 180 and 73 kDa subunits of DNA polymerase alpha as well as that for encoding PCNA. In this study, we found two sequences homologous to DRE (5'-TATCGATA-3') in the 5'-flanking region (-370 to -357 with respect to the transcription initiation site) of the D-raf gene and confirmed transcriptional activity through gel mobility shift assays, transient CAT assays, and spatial patterns of lacZ expression in transgenic larval tissues carrying D-raf and lacZ fusion genes. Further, we demonstrated that the D-raf gene is another target of the Zerknüllt (Zen) protein with observation of D-raf repression by Zen protein in cultured cells and its ectopic expression in the dorsal region of the homozygous zen mutant embryo. The evidence of DRE/DREF involvement in regulation of the D-raf gene obtained in this study strongly supports the idea that the DRE/DREF system is responsible for the coordinated regulation of cell proliferation-related genes in Drosophila.
Collapse
Affiliation(s)
- J R Ryu
- Department of Molecular Biology, College of Natural Science, Pusan National University, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Irie A, Kijima H, Ohkawa T, Bouffard DY, Suzuki T, Curcio LD, Holm PS, Sassani A, Scanlon KJ. Anti-oncogene ribozymes for cancer gene therapy. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1997; 40:207-57. [PMID: 9217927 DOI: 10.1016/s1054-3589(08)60141-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- A Irie
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California 91010, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Alessandrini A, Greulich H, Huang W, Erikson RL. Mek1 phosphorylation site mutants activate Raf-1 in NIH 3T3 cells. J Biol Chem 1996; 271:31612-8. [PMID: 8940180 DOI: 10.1074/jbc.271.49.31612] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
MAP (mitogen-activated protein) kinases are activated by a family of dual specificity kinases called Meks (MAP kinase/Erk kinase). Mek1 can be activated by Raf by phosphorylation on serine 218 and serine 222. Mutation of these sites to acidic residues leads to constitutively active Mek1 in some cases. When fibroblast lines were infected with high titer retroviral stocks carrying these Mek1 genes, the resultant transformation and morphological changes correlated with the kinase activity of the respective Mek1 enzymes. Although [Asp218]- and [Asp218,Asp222]Mek immunoprecipitated from clonal cell lines could phosphorylate kinase-inactive Erk1 equally well in vitro, the endogenous MAP kinase activity was 5-7-fold greater in [Asp218]Mek1-infected clonal lines, and did not correlate with the degree of transformation. Analysis of the Erk1 pathway revealed Raf-1 activation, which correlated qualitatively with the MAP kinase activity seen in the [Asp218]- and [Asp218,Asp222]Mek1-infected clonal cell lines. Expression of dominant negative Ras did not affect the elevated Raf-1 activity observed in these cells, however. These data suggest that Mek1 phosphorylation site mutants activate Raf-1 and MAP kinase by a Ras-independent pathway and that the mechanism by which transformation occurs may utilize pathways that are MAP kinase-independent.
Collapse
Affiliation(s)
- A Alessandrini
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | | | | | | |
Collapse
|
42
|
Widelitz RB, Jiang TX, Noveen A, Chen CW, Chuong CM. FGF induces new feather buds from developing avian skin. J Invest Dermatol 1996; 107:797-803. [PMID: 8941663 DOI: 10.1111/1523-1747.ep12330553] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Induction of skin appendages involves a cascade of molecular events. The fibroblast growth factor (FGF) family of peptide growth factors is involved in cell proliferation and morphogenesis. We explored the role of the FGFs during skin appendage induction using developing chicken feather buds as a model. FGF-1, FGF-2, or FGF-4 was added directly to the culture medium or was released from pre-soaked Affigel blue beads. Near the midline, FGFs led to fusion of developing feather buds, representing FGFs' ability to expand feather bud domains in developing skin. In lateral regions of the explant where feather placodes have not formed, FGF treatment produces a zone of condensation and a region with an increased number of feather buds. In ventral epidermis that is normally apteric (without feathers), FGFs can also induce new feather buds. Like normal feather buds, the newly induced buds express Shh. The expression of Grb, Ras, Raf, and Erk, intracellular signaling molecules known to be downstream to tyrosine kinase receptors such as the FGF receptor, was enriched in feather bud domains. Genistein, an inhibitor of tyrosine kinase, suppressed feather bud formation and the effect of FGF. These results indicate that there are varied responses to FGFs depending on epithelial competence. All the phenotypic responses, however, show that FGFs facilitate the formation of skin appendage domains.
Collapse
Affiliation(s)
- R B Widelitz
- Department of Pathology, University of Southern California, Los Angeles 90033, USA
| | | | | | | | | |
Collapse
|
43
|
Xia K, Mukhopadhyay NK, Inhorn RC, Barber DL, Rose PE, Lee RS, Narsimhan RP, D'Andrea AD, Griffin JD, Roberts TM. The cytokine-activated tyrosine kinase JAK2 activates Raf-1 in a p21ras-dependent manner. Proc Natl Acad Sci U S A 1996; 93:11681-6. [PMID: 8876196 PMCID: PMC38118 DOI: 10.1073/pnas.93.21.11681] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
JAK2, a member of the Janus kinase superfamily was found to interact functionally with Raf-1, a central component of the ras/mitogen-activated protein kinase signal transduction pathway. Interferon-gamma and several other cytokines that are known to activate JAK2 kinase were also found to stimulate Raf-1 kinase activity toward MEK-1 in mammalian cells. In the baculovirus coexpression system, Raf-1 was activated by JAK2 in the presence of p21ras. Under these conditions, a ternary complex of p21ras, JAK2, and Raf-1 was observed. In contrast, in the absence of p21ras, coexpression of JAK2 and Raf-1 resulted in an overall decrease in the Raf-1 kinase activity. In addition, JAK2 phosphorylated Raf-1 at sites different from those phosphorylated by pp60v-src. In mammalian cells treated with either erythropoietin or interferon-gamma, a small fraction of Raf-1 coimmunoprecipitated with JAK2 in lysates of cells in which JAK2 was activated as judged by its state of tyrosine phosphorylation. Taken together, these data suggest that JAK2 and p21ras cooperate to activate Raf-1.
Collapse
Affiliation(s)
- K Xia
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Geilen CC, Wieprecht M, Orfanos CE. The mitogen-activated protein kinases system (MAP kinase cascade): its role in skin signal transduction. A review. J Dermatol Sci 1996; 12:255-62. [PMID: 8884531 DOI: 10.1016/0923-1811(95)00481-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mitogen-activated protein (MAP) kinases are proline-directed kinases which are downstream components of a pathway involving p21ras and the serine/threonine kinase Raf-1. They represent an important link between the signal transduction processes at the level of the plasma membrane and the final nuclear events. Not only various growth factors and cytokines, but also other signals such as UV-light or extracellular matrix components are able to activate MAP kinases. We believe that the MAP kinase cascade may play a significant role in regulating cell proliferation and differentiation in human epidermis. In this review we summarize the rapidly increasing knowledge in this field of signal transduction and discuss some very recent results on MAP kinases and their role in skin biology.
Collapse
Affiliation(s)
- C C Geilen
- Department of Dermatology, University Medical Center Benjamin Franklin, Free University of Berlin, Germany
| | | | | |
Collapse
|
45
|
Meffert S, Stoll M, Steckelings UM, Bottari SP, Unger T. The angiotensin II AT2 receptor inhibits proliferation and promotes differentiation in PC12W cells. Mol Cell Endocrinol 1996; 122:59-67. [PMID: 8898348 DOI: 10.1016/0303-7207(96)03873-7] [Citation(s) in RCA: 168] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Angiotensin II (ANG II) has been implicated in cell growth and differentiation. We investigated the effect of AT2 receptor stimulation on proliferation and morphological differentiation in cells of neuronal origin by using the pheochromocytoma derived cell line, PC12W. ANG II (10(-8)-10(-6) M) inhibited fetal calf serum (FCS)-induced cell proliferation in a concentration dependent manner. In half of the experiments, the epidermal growth factor (EGF) exerted a mitogenic action which was concentration-dependently inhibited by ANG II. In the other half of the experiments, EGF had an antimitogenic effect which was further enhanced by ANG II (maximally at 10(-6) M). Treatment with nerve growth factor (NGF) induced an inhibition of [3H]thymidine incorporation, which was enhanced by ANG II, maximally 25% at the highest concentration. The effects of ANG II on [3H]thymidine incorporation were reflected by those on cell number and were prevented by the AT2 receptor antagonist, PD123177, but not influenced by the AT1 receptor antagonist, losartan. The ANG II-induced inhibition of cell proliferation was paralleled by morphological differentiation in response to daily treatment with ANG II. ANG II also enhanced low-dose NGF-induced neurite formation. Again, these effects of ANG II were abolished by the AT2 receptor antagonist, PD123177. Our data in PC12W cells show that the AT2 receptor not only inhibits growth factor-induced proliferation and enhances the NGF-mediated growth arrest but also induces morphological differentiation in cells of neuronal origin. These findings strongly support the hypothesis that the AT2 receptor promotes differentiation in neuronal cells.
Collapse
Affiliation(s)
- S Meffert
- Department of Pharmacology, Christian-Albrechts-University, Kiel, Germany
| | | | | | | | | |
Collapse
|
46
|
Khosravi-Far R, White MA, Westwick JK, Solski PA, Chrzanowska-Wodnicka M, Van Aelst L, Wigler MH, Der CJ. Oncogenic Ras activation of Raf/mitogen-activated protein kinase-independent pathways is sufficient to cause tumorigenic transformation. Mol Cell Biol 1996; 16:3923-33. [PMID: 8668210 PMCID: PMC231389 DOI: 10.1128/mcb.16.7.3923] [Citation(s) in RCA: 302] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Substantial evidence supports a critical role for the activation of the Raf-1/MEK/mitogen-activated protein kinase pathway in oncogenic Ras-mediated transformation. For example, dominant negative mutants of Raf-1, MEK, and mitogen-activated protein kinase all inhibit Ras transformation. Furthermore, the observation that plasma membrane-localized Raf-1 exhibits the same transforming potency as oncogenic Ras suggests that Raf-1 activation alone is sufficient to mediate full Ras transforming activity. However, the recent identification of other candidate Ras effectors (e.g., RalGDS and phosphatidylinositol-3 kinase) suggests that activation of other downstream effector-mediated signaling pathways may also mediate Ras transforming activity. In support of this, two H-Ras effector domain mutants, H-Ras(12V, 37G) and H-Ras(12V, 40C), which are defective for Raf binding and activation, induced potent tumorigenic transformation of some strains of NIH 3T3 fibroblasts. These Raf-binding defective mutants of H-Ras induced a transformed morphology that was indistinguishable from that induced by activated members of Rho family proteins. Furthermore, the transforming activities of both of these mutants were synergistically enhanced by activated Raf-1 and inhibited by the dominant negative RhoA(19N) mutant, indicating that Ras may cause transformation that occurs via coordinate activation of Raf-dependent and -independent pathways that involves Rho family proteins. Finally, cotransfection of H-Ras(12V, 37G) and H-Ras(12V, 40C) resulted in synergistic cooperation of their focus-forming activities, indicating that Ras activates at least two Raf-independent, Ras effector-mediated signaling events.
Collapse
Affiliation(s)
- R Khosravi-Far
- Department of Pharmacology, University of North Carolina at Chapel Hill, 27599-7365,USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
The activation of the serine/threonine kinase, Raf-1, serves to connect upstream protein tyrosine kinases to downstream signaling events. We previously reported that FcgammaRI stimulation of interferon gamma-differentiated U937 cells (termed U937IF cells) induces a mobility shift in Erk2. Herein, we report that cross-linking of FcgammaRI receptor in U937IF cells induces a marked tyrosine phosphorylation of Raf-1 (10-fold increase). Tyrosine phosphorylation of Raf-1 is induced by FcgammaRI activation and not by PMA (1 microg/ml), N-formyl-Met-Leu-Phe (1 microM), calcium ionophore (1 microM), thrombin (0.05 unit/ml), FcgammaRII, or FcgammaRIII stimulation. The kinetics of Raf-1 tyrosine phosphorylation is rapid, reaching peak levels 1-2 min after FcgammaRI activation, and the tyrosine phosphorylation of Raf-1 precedes the activation of the respiratory burst. FcgammaRI cross-linking induces the tyrosine phosphorylation of Shc; tyrosine-phosphorylated Shc binds to Grb2 forming a Shc-Grb2 complex. The data provide evidence that the FcgammaRI receptor signals via the upstream activation of nonreceptor protein tyrosine kinases, which leads to the subsequent activation of Ras family GTPases and serine/threonine kinases, Raf-1 and mitogen-activated protein kinase.
Collapse
Affiliation(s)
- R K Park
- Neil Bogart Memorial Laboratories, Division of Hematology-Oncology, Childrens Hospital Los Angeles and University of Southern California School of Medicine, Los Angeles, California 90027, USA
| | | | | |
Collapse
|
48
|
Chen HL, Rudland PS, Smith JA, Fernig DG. Late signals are required for the stimulation of DNA synthesis in rat mammary fibroblasts by growth factors. Biosci Rep 1996; 16:249-63. [PMID: 8842375 DOI: 10.1007/bf01207339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Maximal stimulation of DNA synthesis in quiescent rat mammary (Rama) 27 fibroblasts is elicited by epidermal growth factor (EGF) or basic fibroblast growth factor (bFGF) 18 h after the initial addition of the growth factors-the 'lag' period. At maximally-stimulating concentrations, EGF and bFGF are interchangeable 9 h after their initial addition. When the initial concentration of growth factor is below that required to elicit a maximal response, it is possible to increase the level of DNA synthesis by increasing the concentration of growth factor 9 h after its initial addition. When the initial concentration of growth factor is high, substitution by a lower concentration of growth factor after 9 h allows a greater proportion of cells to synthesize DNA than would be expected from a continuous low dose of growth factor. Similar results are obtained when both the growth factor and its concentration are changed 9 h after the initial addition of growth factor. However, when EGF at a low concentration is substituted for a high concentration of EGF or bFGF the resulting increase in the levels of DNA synthesis is greater when EGF rather than bFGF is added for a second time. The half-life of the growth-stimulatory signals delivered by EGF and by bFGF 9 h after their initial addition is 1-2 h. These results suggest that to stimulate DNA synthesis: (i) EGF or bFGF must deliver a signal(s) continuously; (ii) the initial signals produced by EGF and bFGF are equivalent; (iii) the signals produced between 9-18 h by EGF may be different to those produced by bFGF.
Collapse
Affiliation(s)
- H L Chen
- Department of Biochemistry, University of Liverpool, UK
| | | | | | | |
Collapse
|
49
|
Gorman C, Skinner RH, Skelly JV, Neidle S, Lowe PN. Equilibrium and kinetic measurements reveal rapidly reversible binding of Ras to Raf. J Biol Chem 1996; 271:6713-9. [PMID: 8636091 DOI: 10.1074/jbc.271.12.6713] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Raf is a serine/threonine kinase that binds through its amino-terminal regulatory domain to the GTP form of Ras and thereby activates the mitogen-activated protein kinase pathway. In this study, we have characterized the interaction of the Ras-binding domain of Raf with Ras using equilibrium binding methods (scintillation proximity assay and fluorescence anisotropy), rather than with more widely used nonequilibrium procedures (such as enzyme-linked immunosorbent assay and affinity precipitation). Initial studies using glutathione S-transferase fusion proteins with either residues 1-257 or 1-190 of Raf showed that although it was possible to detect Ras binding using an enzyme-linked immunosorbent assay or affinity precipitation, it was substoichiometric; under equilibrium conditions with only a small excess of Raf almost no binding was detected. This difference was probably due to the presence of a high percentage of inactive Raf protein. Further studies used protein containing residues 51-131 of Raf, which expressed in Escherichia coli as a stable glutathione S-transferase fusion. With this protein, binding with Ras could readily be measured under equilibrium conditions. The catalytic domain of neurofibromin inhibited binding of Ras to Raf, and Raf inhibited the binding of Ras to neurofibromin showing that Raf and neurofibromin cannot be bound simultaneously to Ras. The affinities of interaction of neurofibromin and Raf with Harvey-RasLeu-61 were similar. The rate constant for dissociation of Raf from Ras was estimated to be >1 min-1, suggesting that Ras, Raf, and neurofibromin may be in rapid equilibrium in the cell. In contrast to previous reports, under equilibrium conditions there was no evidence for a difference in affinity between the minimal Ras binding domain of Raf (residues 51-131) and a region containing an additional 16 carboxyl-terminal amino acids, suggesting that residues 132-147 do not form a critical binding determinant.
Collapse
Affiliation(s)
- C Gorman
- Wellcome Research Laboratories, Langley Court, South Eden Park Road, Beckenham, Kent BR3 3BS, United Kingdom
| | | | | | | | | |
Collapse
|
50
|
Manganiello VC, Degerman E, Taira M, Kono T, Belfrage P. Type III cyclic nucleotide phosphodiesterases and insulin action. CURRENT TOPICS IN CELLULAR REGULATION 1996; 34:63-100. [PMID: 8646851 DOI: 10.1016/s0070-2137(96)80003-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- V C Manganiello
- Laboratory of Cellular Metabolism, NHLBI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|