1
|
Ramsey A, Akana L, Miyajima E, Douglas S, Gray J, Rowland A, Sharma KD, Xu J, Xie JY, Zhou GL. CAP1 (cyclase-associated protein 1) mediates the cyclic AMP signals that activate Rap1 in stimulating matrix adhesion of colon cancer cells. Cell Signal 2023; 104:110589. [PMID: 36621727 PMCID: PMC9908859 DOI: 10.1016/j.cellsig.2023.110589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
We previously reported that CAP1 (Cyclase-Associated Protein 1) regulates matrix adhesion in mammalian cells through FAK (Focal Adhesion Kinase). More recently, we discovered a phosphor-regulation mechanism for CAP1 through the Ser307/Ser309 tandem site that is of critical importance for all CAP1 functions. However, molecular mechanisms underlying the CAP1 function in adhesion and its regulation remain largely unknown. Here we report that Rap1 also facilitates the CAP1 function in adhesion, and more importantly, we identify a novel signaling pathway where CAP1 mediates the cAMP signals, through the cAMP effectors Epac (Exchange proteins directly activated by cAMP) and PKA (Protein Kinase A), to activate Rap1 in stimulating matrix adhesion in colon cancer cells. Knockdown of CAP1 led to opposite adhesion phenotypes in SW480 and HCT116 colon cancer cells, with reduced matrix adhesion and reduced FAK and Rap1 activities in SW480 cells while it stimulated matrix adhesion as well as FAK and Rap1 activities in HCT116 cells. Importantly, depletion of CAP1 abolished the stimulatory effects of the cAMP activators forskolin and isoproterenol, as well as that of Epac and PKA, on matrix adhesion in both cell types. Our results consistently support a required role for CAP1 in the cAMP activation of Rap1. Identification of the key role for CAP1 in linking the major second messenger cAMP to activation of Rap1 in stimulating adhesion, which may potentially also regulate proliferation in other cell types, not only vertically extends our knowledge on CAP biology, but also carries important translational potential for targeting CAP1 in cancer therapeutics.
Collapse
Affiliation(s)
- Auburn Ramsey
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Lokesh Akana
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Erina Miyajima
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Spencer Douglas
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Joshua Gray
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Alyssa Rowland
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Krishna Deo Sharma
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA
| | - Jianfeng Xu
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA; Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA; College of Agriculture, Arkansas State University, State University, AR 72467, USA
| | - Jennifer Y Xie
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA; Department of Basic Sciences, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR 72401, USA
| | - Guo-Lei Zhou
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA; Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA.
| |
Collapse
|
2
|
Peng Q, Guo R, Zhou Y, Teng R, Cao Y, Mu S. Comparison of Gelatin/Polylysine- and Silk Fibroin/SDF-1α-Coated Mesenchymal Stem Cell-Seeded Intracranial Stents. Macromol Biosci 2022; 23:e2200402. [PMID: 36541928 DOI: 10.1002/mabi.202200402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Endothelialization of the aneurysmal neck is essential for aneurysm healing after endovascular treatment. Mesenchymal stem cell (MSC)-seeded stents can promote aneurysm repair. The biological effects of coated and uncoated nitinol intracranial stents seeded with MSCs on vascular cells and macrophage proliferation and inflammation are investigated. Two stent coatings that exert pro-aggregation effects on MSCs via different mechanisms are examined: gelatin/polylysine (G/PLL), which enhances cell adhesion, and silk fibroin/SDF-1α (SF/SDF-1α), which enhances chemotaxis. The aim is to explore the feasibility of MSC-seeded coated stents in the treatment of intracranial aneurysms. The G/PLL coating provides the highest cytocompatibility and blood compatibility substrate for MSCs and vascular cells and promotes cell adhesion and proliferation. Moreover, it enhances MSC secretion and regulation of vascular cell and macrophage proliferation and chemotaxis. Although the SF/SDF-1α coating promotes MSC secretion and vascular cell chemotaxis, it induces a greater degree of macrophage proliferation, chemotaxis, and secretion of pro-inflammatory factors. MSC-seeded stents coated with G/PLL may benefit stent surface endothelialization and reduce the inflammatory response after endovascular treatment of intracranial aneurysm. These effects may improve aneurysm healing and increase the cure rate.
Collapse
Affiliation(s)
- Qichen Peng
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Ruimin Guo
- Healthina Academy of Biomedicine, Tianjin Economic-Technological Development Area, HAB-TEDA, Tianjin, 300457, China.,Tangyi holdings (Shenzhen) Co., LTD, Shenzhen, 518101, China
| | - Yangyang Zhou
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Ruidi Teng
- Healthina Academy of Biomedicine, Tianjin Economic-Technological Development Area, HAB-TEDA, Tianjin, 300457, China.,Tangyi holdings (Shenzhen) Co., LTD, Shenzhen, 518101, China
| | - Yulin Cao
- Healthina Academy of Biomedicine, Tianjin Economic-Technological Development Area, HAB-TEDA, Tianjin, 300457, China.,Tangyi holdings (Shenzhen) Co., LTD, Shenzhen, 518101, China
| | - Shiqing Mu
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| |
Collapse
|
3
|
Hinton TJ, Jallerat Q, Palchesko RN, Park JH, Grodzicki MS, Shue HJ, Ramadan MH, Hudson AR, Feinberg AW. Three-dimensional printing of complex biological structures by freeform reversible embedding of suspended hydrogels. SCIENCE ADVANCES 2015; 1:e1500758. [PMID: 26601312 PMCID: PMC4646826 DOI: 10.1126/sciadv.1500758] [Citation(s) in RCA: 1022] [Impact Index Per Article: 102.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/02/2015] [Indexed: 05/17/2023]
Abstract
We demonstrate the additive manufacturing of complex three-dimensional (3D) biological structures using soft protein and polysaccharide hydrogels that are challenging or impossible to create using traditional fabrication approaches. These structures are built by embedding the printed hydrogel within a secondary hydrogel that serves as a temporary, thermoreversible, and biocompatible support. This process, termed freeform reversible embedding of suspended hydrogels, enables 3D printing of hydrated materials with an elastic modulus <500 kPa including alginate, collagen, and fibrin. Computer-aided design models of 3D optical, computed tomography, and magnetic resonance imaging data were 3D printed at a resolution of ~200 μm and at low cost by leveraging open-source hardware and software tools. Proof-of-concept structures based on femurs, branched coronary arteries, trabeculated embryonic hearts, and human brains were mechanically robust and recreated complex 3D internal and external anatomical architectures.
Collapse
Affiliation(s)
- Thomas J. Hinton
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Quentin Jallerat
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Rachelle N. Palchesko
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Joon Hyung Park
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Martin S. Grodzicki
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Hao-Jan Shue
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Mohamed H. Ramadan
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Andrew R. Hudson
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Adam W. Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Corresponding author. E-mail:
| |
Collapse
|
4
|
Vicenova M, Nechvatalova K, Chlebova K, Kucerova Z, Leva L, Stepanova H, Faldyna M. Evaluation of in vitro and in vivo anti-inflammatory activity of biologically active phospholipids with anti-neoplastic potential in porcine model. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:339. [PMID: 25234616 PMCID: PMC4179840 DOI: 10.1186/1472-6882-14-339] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 09/11/2014] [Indexed: 12/11/2022]
Abstract
Background This study aims to investigate the anti-inflammatory effect of biologically active phospholipids (BAP) used in preparations for clinical practice in humans. Until date, except anti-neoplastic ability, little is known about anti-inflammatory property of the phospholipids. Methods While the course of bacterially induced acute pneumonia and markers of inflammation were studied in in vivo system in pigs orally supplemented with BAP, the pro- and anti-inflammatory response of lipopolysaccharide-stimulated porcine monocyte-derived macrophages to 24 h- and 48 h-treatmeant by BAP was investigated in in vitro system. In vivo, the animal health status was monitored and pro-inflammatory IL-1β and IL-8 in sera were detected by ELISA during the experiment, while bronchoalveolar lavage fluids (BALF) and the lungs were examined post-mortem. Total and differential counts of white blood cell (WBC) were determined in blood and BALF. In vitro, mRNA expression of pro-inflammatory (TNF-α, IL-1β, CXCL10) and anti-inflammatory (IL-10 and Arg1) cytokines, and level of activated caspase 1 and phosphorylated protein kinase C epsilon (pPKCϵ), were studied using qRT-PCR and Western blot, respectively. For the purposes of both systems, 6 animals were used in each of the BAP-supplemented and the control groups. Results In vivo, BAP had a positive influence on the course of the disease. The immunomodulatory effects of BAP were confirmed by lower levels of IL-1β, IL-8, and a lower WBC count in the supplemented group in comparison with the control group. A lower percentage of lung parenchyma was affected in the supplemented group comparing to the control group (on average, 4% and 34% of tissue, respectively). In vitro, BAP suppressed mRNA expression of mRNA for IL-10 and all pro-inflammatory cytokines tested. This down-regulation was dose- and time-dependent. Arg1 mRNA expression remained unaffected. Further dose- and time-dependent suppression of the activated caspase 1 and pPKCϵ was detected in macrophages when treated with BAP. Conclusions Our results demonstrate that BAP has anti-inflammatory and immunomodulatory properties, thus emphasizing the potential of this compound as a natural healing agent.
Collapse
|
5
|
Ong ST, Freeley M, Skubis-Zegadło J, Fazil MHUT, Kelleher D, Fresser F, Baier G, Verma NK, Long A. Phosphorylation of Rab5a protein by protein kinase Cϵ is crucial for T-cell migration. J Biol Chem 2014; 289:19420-34. [PMID: 24872409 DOI: 10.1074/jbc.m113.545863] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Rab GTPases control membrane traffic and receptor-mediated endocytosis. Within this context, Rab5a plays an important role in the spatial regulation of intracellular transport and signal transduction processes. Here, we report a previously uncharacterized role for Rab5a in the regulation of T-cell motility. We show that Rab5a physically associates with protein kinase Cϵ (PKCϵ) in migrating T-cells. After stimulation of T-cells through the integrin LFA-1 or the chemokine receptor CXCR4, Rab5a is phosphorylated on an N-terminal Thr-7 site by PKCϵ. Both Rab5a and PKCϵ dynamically interact at the centrosomal region of migrating cells, and PKCϵ-mediated phosphorylation on Thr-7 regulates Rab5a trafficking to the cell leading edge. Furthermore, we demonstrate that Rab5a Thr-7 phosphorylation is functionally necessary for Rac1 activation, actin rearrangement, and T-cell motility. We present a novel mechanism by which a PKCϵ-Rab5a-Rac1 axis regulates cytoskeleton remodeling and T-cell migration, both of which are central for the adaptive immune response.
Collapse
Affiliation(s)
- Seow Theng Ong
- From the From the Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Michael Freeley
- From the From the Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Joanna Skubis-Zegadło
- From the From the Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland, Department of Applied Pharmacy and Bioengineering, Medical University of Warsaw, 02-091 Warsaw, Poland
| | | | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553, Faculty of Medicine, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom, and
| | - Friedrich Fresser
- the Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, A-6020 Innsbruck, Austria
| | - Gottfried Baier
- the Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, A-6020 Innsbruck, Austria
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553,
| | - Aideen Long
- From the From the Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland,
| |
Collapse
|
6
|
Jain K, Basu A. The Multifunctional Protein Kinase C-ε in Cancer Development and Progression. Cancers (Basel) 2014; 6:860-78. [PMID: 24727247 PMCID: PMC4074807 DOI: 10.3390/cancers6020860] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 03/27/2014] [Accepted: 04/01/2014] [Indexed: 12/31/2022] Open
Abstract
The protein kinase C (PKC) family proteins are important signal transducers and have long been the focus of cancer research. PKCɛ, a member of this family, is overexpressed in most solid tumors and plays critical roles in different processes that lead to cancer development. Studies using cell lines and animal models demonstrated the transforming potential of PKCɛ. While earlier research established the survival functions of PKCɛ, recent studies revealed its role in cell migration, invasion and cancer metastasis. PKCɛ has also been implicated in epithelial to mesenchymal transition (EMT), which may be the underlying mechanism by which it contributes to cell motility. In addition, PKCɛ affects cell-extracellular matrix (ECM) interactions by direct regulation of the cytoskeletal elements. Recent studies have also linked PKCɛ signaling to cancer stem cell functioning. This review focuses on the role of PKCɛ in different processes that lead to cancer development and progression. We also discussed current literatures on the pursuit of PKCɛ as a target for cancer therapy.
Collapse
Affiliation(s)
- Kirti Jain
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Institute for Cancer Research, and Focused on Resources for her Health Education and Research, Fort Worth, TX 76107, USA.
| | - Alakananda Basu
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Institute for Cancer Research, and Focused on Resources for her Health Education and Research, Fort Worth, TX 76107, USA.
| |
Collapse
|
7
|
Integrin and GPCR Crosstalk in the Regulation of ASM Contraction Signaling in Asthma. J Allergy (Cairo) 2012; 2012:341282. [PMID: 23056062 PMCID: PMC3465959 DOI: 10.1155/2012/341282] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 07/24/2012] [Indexed: 12/15/2022] Open
Abstract
Airway hyperresponsiveness (AHR) is one of the cardinal features of asthma. Contraction of airway smooth muscle (ASM) cells that line the airway wall is thought to influence aspects of AHR, resulting in excessive narrowing or occlusion of the airway. ASM contraction is primarily controlled by agonists that bind G protein-coupled receptor (GPCR), which are expressed on ASM. Integrins also play a role in regulating ASM contraction signaling. As therapies for asthma are based on symptom relief, better understanding of the crosstalk between GPCRs and integrins holds good promise for the design of more effective therapies that target the underlying cellular and molecular mechanism that governs AHR. In this paper, we will review current knowledge about integrins and GPCRs in their regulation of ASM contraction signaling and discuss the emerging concept of crosstalk between the two and the implication of this crosstalk on the development of agents that target AHR.
Collapse
|
8
|
Totoń E, Ignatowicz E, Skrzeczkowska K, Rybczyńska M. Protein kinase Cε as a cancer marker and target for anticancer therapy. Pharmacol Rep 2011; 63:19-29. [DOI: 10.1016/s1734-1140(11)70395-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 06/10/2010] [Indexed: 01/23/2023]
|
9
|
Brenner W, Beitz S, Schneider E, Benzing F, Unger RE, Roos FC, Thüroff JW, Hampel C. Adhesion of renal carcinoma cells to endothelial cells depends on PKCmu. BMC Cancer 2010; 10:183. [PMID: 20459627 PMCID: PMC2873397 DOI: 10.1186/1471-2407-10-183] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 05/06/2010] [Indexed: 11/29/2022] Open
Abstract
Background The formation of metastases includes the separation of tumor cells from the primary tumor, cell migration into subendothelial tissue and cell proliferation in secondary organ. In this process, cell adhesion of tumor cells to the endothelium is an essential requirement for formation of metastases. Protein kinase C (PKC) regulates adhesion and proliferation. To identify a relation between PKC isoforms and tumor progression in renal cell carcinoma (RCC), the influence of PKC isoforms on cell adhesion and proliferation, and possible influences of integrins were analyzed in RCC cells. Methods The experiments were performed in the RCC cell lines CCF-RC1 and CCF-RC2 after pre-incubation (16 h) with the PKC inhibitors GF109203X (inhibits PKCα, βI, βII, γ, δ and ε), GÖ6976 (inhibits PKCα, βI and μ), RO31-8220 (inhibits PKCα, βI, βII, γ and ε) and rottlerin (inhibits PKCδ). Cell adhesion was assessed through adherence of RCC cells to an endothelial monolayer. Cell proliferation was analyzed by a BrdU incorporation assay. The expression of β1 integrins was analyzed by flow cytometry. Results In CCF-RC1 cells, cell adhesion was significantly reduced by GÖ6976 to 55% and by RO31-8220 to 45% of control. In CCF-RC2 cells, only GÖ6976 induced a significant reduction of cell adhesion to 50% of control levels. Proliferation of both cell lines was reduced by rottlerin to 39% and 45% of control, respectively. The β1 integrin expression on the cell surface of CCF-RC1 and CCR-RC2 cells was decreased by RO31-8220 to 8% and 7% of control, respectively. β2 and β3 integrins were undetectable in both cell lines. Conclusions The combination of the PKC inhibitors leads to the assumption that PKCμ influences cell adhesion in CCF-RC1 and CCF-RC2 cells, whereas in CCF-RC1 cells PKCε also seems to be involved in this process. The expression of β1 integrins appears to be regulated in particular by PKCε. Cell proliferation was inhibited by rottlerin, so that PKCδ might be involved in cell proliferation in these cells.
Collapse
Affiliation(s)
- Walburgis Brenner
- Department of Urology, University Medical Center Mainz, Mainz, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Fortin S, Le Mercier M, Camby I, Spiegl-Kreinecker S, Berger W, Lefranc F, Kiss R. Galectin-1 is implicated in the protein kinase C epsilon/vimentin-controlled trafficking of integrin-beta1 in glioblastoma cells. Brain Pathol 2009; 20:39-49. [PMID: 18947333 PMCID: PMC2805865 DOI: 10.1111/j.1750-3639.2008.00227.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cell motility and resistance to apoptosis characterize glioblastoma (GBM) growth and malignancy. In our current work we report that galectin‐1, a homodimeric adhesion molecule and carbohydrate‐binding protein with affinity for β‐galactosides, is linked with cell surface expression of integrin β1 and the process of integrin trafficking. Using immunofluorescence, depletion of galectin‐1 through both stable knockdown and transient‐targeted small interfering RNA (siRNA) treatment induces an intracellular accumulation of integrin‐β1 coincident with a diminution of integrin‐β1 at points of cellular adhesion at the cell membrane. Galectin‐1 depletion does not alter the gene expression level of integrin‐β1. Transient galectin‐1 depletion effectuates as well the perinuclear accumulation of protein kinase C epsilon (PKCε) and the intermediate filament vimentin, both of which have been shown to mediate integrin recycling in motile cells. Our results argue for the involvement of galectin‐1 in the PKCε/vimentin‐controlled trafficking of integrin‐β1. The understanding of molecular mediators such as galectin‐1 and the pathways through which they drive the cell invasion so descriptive of GBM is anticipated to reveal potential therapeutic targets that promote glioma malignancy.
Collapse
Affiliation(s)
- Shannon Fortin
- Laboratory of Toxicology, Institute of Pharmacy, Univesité Libre de Bruxelles, Brussels
| | | | | | | | | | | | | |
Collapse
|
11
|
Signal transduction of constitutively active protein kinase C epsilon. Cell Signal 2009; 21:745-52. [DOI: 10.1016/j.cellsig.2009.01.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Accepted: 01/03/2009] [Indexed: 11/18/2022]
|
12
|
Meloni I, Parri V, De Filippis R, Ariani F, Artuso R, Bruttini M, Katzaki E, Longo I, Mari F, Bellan C, Dotti CG, Renieri A. The XLMR gene ACSL4 plays a role in dendritic spine architecture. Neuroscience 2009; 159:657-69. [PMID: 19166906 DOI: 10.1016/j.neuroscience.2008.11.056] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 11/21/2008] [Accepted: 11/22/2008] [Indexed: 12/13/2022]
Abstract
ACSL4 is a gene involved in non-syndromic X-linked mental retardation. It encodes for a ubiquitous protein that adds coenzyme A to long-chain fatty acids, with a high substrate preference for arachidonic acid. It presents also a brain-specific isoform deriving from an alternative splicing and containing 41 additional N-terminal amino acids. To start to unravelling the link between ACSL4 and mental retardation, we have performed molecular and cell biological studies. By retro-transcription polymerase chain reaction analyses we identified a new transcript with a shorter 5'-UTR region. By immunofluorescence microscopy in embryonic rat hippocampal neurons we report that ACSL4 is associated preferentially to endoplasmic reticulum tubules. ACSL4 knockdown by siRNAs in hippocampal neurons indicated that this protein is largely dispensable for these cells' gross architectural features (i.e. axonal and dendritic formation and final length) yet it is required for the presence of normal spines. In fact, reduced levels of ACSL4 led to a significant reduction in dendritic spine density and an alteration in spine/filopodia distribution. The possible mechanisms behind this phenotype are discussed.
Collapse
Affiliation(s)
- I Meloni
- Medical Genetics Unit, Department of Molecular Biology, University of Siena, Policlinico Le Scotte, Viale Bracci 2, 53100 Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Gorin MA, Pan Q. Protein kinase C epsilon: an oncogene and emerging tumor biomarker. Mol Cancer 2009; 8:9. [PMID: 19228372 PMCID: PMC2647895 DOI: 10.1186/1476-4598-8-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 02/19/2009] [Indexed: 12/30/2022] Open
Abstract
Members of the protein kinase C (PKC) family have long been studied for their contributions to oncogenesis. Among the ten different isoforms of this family of serine/threonine kinases, protein kinase Cε (PKCε) is one of the best understood for its role as a transforming oncogene. In vitro, overexpression of PKCε has been demonstrated to increase proliferation, motility, and invasion of fibroblasts or immortalized epithelial cells. In addition, xenograft and transgenic animal models have clearly shown that overexpression of PKCε is tumorigenic resulting in metastatic disease. Perhaps most important in implicating the epsilon isoform in oncogenesis, PKCε has been found to be overexpressed in tumor-derived cell lines and histopathological tumor specimens from various organ sites. Combined, this body of work provides substantial evidence implicating PKCε as a transforming oncogene that plays a crucial role in establishing an aggressive metastatic phenotype. Reviewed here is the literature that has led to the current understanding of PKCε as an oncogene. Moreover, this review focuses on the PKCε-mediated signaling network for cell motility and explores the interaction of PKCε with three major PKCε signaling nodes: RhoA/C, Stat3 and Akt. Lastly, the emerging role of PKCε as a tumor biomarker is discussed.
Collapse
Affiliation(s)
- Michael A Gorin
- University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| | | |
Collapse
|
14
|
Akita Y. Protein kinase Cε: multiple roles in the function of, and signaling mediated by, the cytoskeleton. FEBS J 2008; 275:3995-4004. [DOI: 10.1111/j.1742-4658.2008.06557.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Breitkreutz D, Braiman-Wiksman L, Daum N, Denning MF, Tennenbaum T. Protein kinase C family: on the crossroads of cell signaling in skin and tumor epithelium. J Cancer Res Clin Oncol 2007; 133:793-808. [PMID: 17661083 DOI: 10.1007/s00432-007-0280-3] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Accepted: 07/03/2007] [Indexed: 12/28/2022]
Abstract
The protein kinase C (PKC) family represents a large group of phospholipid dependent enzymes catalyzing the covalent transfer of phosphate from ATP to serine and threonine residues of proteins. Phosphorylation of the substrate proteins induces a conformational change resulting in modification of their functional properties. The PKC family consists of at least ten members, divided into three subgroups: classical PKCs (alpha, betaI, betaII, gamma), novel PKCs (delta, epsilon, eta, theta), and atypical PKCs (zeta, iota/lambda). The specific cofactor requirements, tissue distribution, and cellular compartmentalization suggest differential functions and fine tuning of specific signaling cascades for each isoform. Thus, specific stimuli can lead to differential responses via isoform specific PKC signaling regulated by their expression, localization, and phosphorylation status in particular biological settings. PKC isoforms are activated by a variety of extracellular signals and, in turn, modify the activities of cellular proteins including receptors, enzymes, cytoskeletal proteins, and transcription factors. Accordingly, the PKC family plays a central role in cellular signal processing. Accumulating data suggest that various PKC isoforms participate in the regulation of cell proliferation, differentiation, survival and death. These findings have enabled identification of abnormalities in PKC isoform function, as they occur in several cancers. Specifically, the initiation of squamous cell carcinoma formation and progression to the malignant phenotype was found to be associated with distinct changes in PKC expression, activation, distribution, and phosphorylation. These studies were recently further extended to transgenic and knockout animals, which allowed a more direct analysis of individual PKC functions. Accordingly, this review is focused on the involvement of PKC in physiology and pathology of the skin.
Collapse
Affiliation(s)
- D Breitkreutz
- Division of Differentiation and Carcinogenesis (A080/A110), German Cancer Research Center (DKFZ), POB 101949, Im Neuenheimer Feld 280, 69009, Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
16
|
Tsubaki M, Matsuoka H, Yamamoto C, Kato C, Ogaki M, Satou T, Itoh T, Kusunoki T, Tanimori Y, Nishida S. The protein kinase C inhibitor, H7, inhibits tumor cell invasion and metastasis in mouse melanoma via suppression of ERK1/2. Clin Exp Metastasis 2007; 24:431-8. [PMID: 17636410 DOI: 10.1007/s10585-007-9080-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Accepted: 05/26/2007] [Indexed: 12/14/2022]
Abstract
Protein kinase C (PKC) has been shown to be a signal transducer during tumorigenesis, tumor cell invasion, and metastasis. Recent studies have reported that the PKC inhibitor, 7-hydroxystaurosporine, inhibits tumor cell invasion. However, the molecular mechanisms of this inhibition of invasion and metastasis are not well understood. In the present study, we attempt to clarify the mechanism by which H7, a PKC inhibitor, inhibits tumor cell invasion and metastasis in the melanoma cell line B16BL6. It was found that H7 inhibits B16BL6 cell invasion and metastasis. We also observed that H7 inhibits the mRNA expression and protein activities of matrix metalloproteinase (MMP)-1, -2, -9 and MT1-MMP. Furthermore, H7 suppresses phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2). However, other signal transduction factors, such as p38 mitogen-activated protein kinase (p38MAPK) and c-Jun N-terminal kinase 1/2 (JNK1/2), were unaffected. Moreover, U0126, a MEK1/2 inhibitor, also inhibited B16BL6 cell invasion and metastasis, as well as the mRNA expression and protein activities of MMP-1, -2, -9 and MT1-MMP. This indicates that H7 inhibits signal transduction through the PKC/MEK/ERK pathway, thereby inhibiting B16BL6 cell invasion and metastasis. These results suggest that PKC inhibitors have potential clinical applications in the treatment of tumor cell metastasis.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kinki University School of Pharmaceutical Sciences, Kowakae, Higashi-Osaka 577-8502, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Slager RE, Sisson JH, Pavlik JA, Johnson JK, Nicolarsen JR, Jerrells TR, Wyatt TA. Inhibition of protein kinase C epsilon causes ciliated bovine bronchial cell detachment. Exp Lung Res 2006; 32:349-62. [PMID: 17090476 PMCID: PMC2100410 DOI: 10.1080/01902140600959630] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
This study defines the in vitro phenomenon of ciliated bovine bronchial epithelial cell (BBEC) detachment from the basal epithelium and regulation of cilia motility mediated through protein kinase C epsilon (PKCepsilon). The authors determined the time course of activation and downregulation of PKCepsilon by the known PKC activator phorbol 12-myristate 13-acetate (PMA) and demonstrate that chemical inhibition of PKC by calphostin C or the novel PKC isoform inhibitor Ro 31-8220 induced striking detachment of ciliated BBECs from the basal cell monolayer within 1 hour, independent of apoptosis or necrotic cell death. The results of this study support a possible novel PKCepsilon-mediated signaling pathway through which ciliated cell attachment is maintained.
Collapse
Affiliation(s)
- Rebecca E Slager
- Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198-5300, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Yosimichi G, Kubota S, Nishida T, Kondo S, Yanagita T, Nakao K, Takano-Yamamoto T, Takigawa M. Roles of PKC, PI3K and JNK in multiple transduction of CCN2/CTGF signals in chondrocytes. Bone 2006; 38:853-63. [PMID: 16431170 DOI: 10.1016/j.bone.2005.11.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Revised: 11/05/2005] [Accepted: 11/11/2005] [Indexed: 10/25/2022]
Abstract
CCN2/connective tissue growth factor (CCN2/CTGF) is known to promote both the proliferation and differentiation of chondrocytes, which actions are mediated by ERK and p38 MAPK, respectively. In this study, we first re-evaluated the involvement of multiple MAPKs therein and found that JNK also mediated such CCN2 signals. Thereafter, we further analyzed the roles of upstream kinases. The involvement of PKC, PI3K and PKA in the CCN2 signaling to promote the maturation, proliferation and terminal differentiation of a human chondrocytic cell line, HCS-2/8 and rabbit primary growth cartilage cells was investigated. As a result, the PKC inhibitor calphostin C repressed all of the effects of CCN2, which were represented by increased synthesis of DNA and proteoglycans and the display of alkaline phosphatase activity. In addition, evaluation of the effect of the PI3K inhibitor wortmannin disclosed the contribution of PI3K in transducing CCN2 signals to promote chondrocyte hypertrophy. This signal was known to be mediated by PKB, which was translocated into the nucleus upon CCN2 stimulation. Of note, calphostin C showed inhibitory effects on the activation of p38 MAPK, ERK and also PKB, whereas it exerted no effect on JNK activation. These results suggest that PKC is a driver of multiple signal transducing kinases that promote the proliferation and differentiation of chondrocytes. The requirement of PI3K in transmitting the signal for terminal differentiation and PKC-independent signaling pathways for the promotion of chondrocytic growth and differentiation, which was mediated by JNK, were also uncovered.
Collapse
Affiliation(s)
- Gen Yosimichi
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Solanum nigrum L. (SNL) has been used in folk medicine for its anti-inflammatory activity. We previously isolated glycoprotein from SNL and observed that it decreased viable HT-29 cell numbers at a low concentration (60 microg/mL). This study investigated the apoptotic signal pathway triggered by glycoprotein isolated from SNL in HT-29 cells. Treatment of HT-29 cells with SNL glycoprotein (60 microg/mL) for 4 hours resulted in a cytotoxic effect of more than 60%, compared with the control. To explain the apoptotic effects of SNL glycoprotein, we investigated its effects on 12-O-tetradecanoylphorbol 13-acetate (TPA)-stimulated protein kinase C (PKC) alpha activity and DNA-binding activity of nuclear factor (NF) kappaB in HT-29 cells, using western blot analysis and electrophoretic mobility shift assays. Results from these experiments showed that SNL glycoprotein has remarkable inhibitory effects on the activities of TPA (100 nM)-stimulated PKCalpha and NF-kappaB in HT-29 cells. They also substantiated that PKCalpha is a part of the TPA-activated upstream signal pathway of NF-kappaB, since NF-kappaB activity was inhibited by staurosporine (a PKC inhibitor) and pyrrolidine dithiocarbamate (an NF-kappaB inhibitor) in a western blot analysis. Furthermore, to verify the triggering of apoptosis by the SNL glycoprotein, we performed DNA fragmentation, nuclear staining, and protein expression assays of apoptotic-related proteins. The amount of DNA fragmentation and apoptotic cell numbers increased in a dose-dependent manner after treatment with SNL glycoprotein. Apoptosis-related protein assays demonstrated that SNL glycoprotein-induced apoptosis is associated with the regulation of bcl-2 and Bax expression. Taken together, the results of this study showed that the activation of PKCalpha, NF-kappaB, and Bax expression by SNL glycoprotein is possibly involved in the apoptotic process. Consequently, these results indicate that SNL glycoprotein causes HT-29 cell death through apoptosis by its ability to modulate anti-apoptotic signals. We suggest that SNL glycoprotein is a natural anti-cancer agent due to its potential to induce apoptosis in HT-29 cells.
Collapse
Affiliation(s)
- Kye-Taek Lim
- #521 Molecular Biochemistry Laboratory, Institute of Biotechnology, Chonnam National University, Yongbong Dong, Kwangju, Korea.
| |
Collapse
|
20
|
Jerdeva GV, Yarber FA, Trousdale MD, Rhodes CJ, Okamoto CT, Dartt DA, Hamm-Alvarez SF. Dominant-negative PKC-epsilon impairs apical actin remodeling in parallel with inhibition of carbachol-stimulated secretion in rabbit lacrimal acini. Am J Physiol Cell Physiol 2005; 289:C1052-68. [PMID: 15930141 PMCID: PMC1414898 DOI: 10.1152/ajpcell.00546.2004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated the involvement of PKC-epsilon in apical actin remodeling in carbachol-stimulated exocytosis in reconstituted rabbit lacrimal acinar cells. Lacrimal acinar PKC-epsilon cosedimented with actin filaments in an actin filament binding assay. Stimulation of acini with carbachol (100 microM, 2-15 min) significantly (P < or = 0.05) increased PKC-epsilon recovery with actin filaments in two distinct biochemical assays, and confocal fluorescence microscopy showed a significant increase in PKC-epsilon association with apical actin in stimulated acini as evidenced by quantitative colocalization analysis. Overexpression of dominant-negative (DN) PKC-epsilon in lacrimal acini with replication-defective adenovirus (Ad) resulted in profound alterations in apical and basolateral actin filaments while significantly inhibiting carbachol-stimulated secretion of bulk protein and beta-hexosaminidase. The chemical inhibitor GF-109203X (10 microM, 3 h), which inhibits PKC-alpha, -beta, -delta, and -epsilon, also elicited more potent inhibition of carbachol-stimulated secretion relative to Gö-6976 (10 microM, 3 h), which inhibits only PKC-alpha and -beta. Transduction of lacrimal acini with Ad encoding syncollin-green fluorescent protein (GFP) resulted in labeling of secretory vesicles that were discharged in response to carbachol stimulation, whereas cotransduction of acini with Ad-DN-PKC-epsilon significantly inhibited carbachol-stimulated release of syncollin-GFP. Carbachol also increased the recovery of secretory component in culture medium, whereas Ad-DN-PKC-epsilon transduction suppressed its carbachol-stimulated release. We propose that DN-PKC-epsilon alters lacrimal acinar apical actin remodeling, leading to inhibition of stimulated exocytosis and transcytosis.
Collapse
Affiliation(s)
| | | | | | - Christopher J. Rhodes
- University of Southern California, Los Angeles CA; Pacific Northwest Research Institute, Seattle WA; and
| | | | | | - Sarah F. Hamm-Alvarez
- Departments of Pharmaceutical Sciences
- Ophthalmology and
- Physiology and Biophysics
- Address correspondence to: Sarah F. Hamm-Alvarez, Ph. D., Department of Pharmaceutical Sciences, USC School of Pharmacy, 1985 Zonal Avenue, Los Angeles CA 90033, 323-442-1445 O, 323-442-1390 F,
| |
Collapse
|
21
|
Dina OA, Hucho T, Yeh J, Malik-Hall M, Reichling DB, Levine JD. Primary afferent second messenger cascades interact with specific integrin subunits in producing inflammatory hyperalgesia. Pain 2005; 115:191-203. [PMID: 15836982 DOI: 10.1016/j.pain.2005.02.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Revised: 02/11/2005] [Accepted: 02/22/2005] [Indexed: 01/19/2023]
Abstract
We recently reported that hyperalgesia induced by the inflammatory mediator prostaglandin E(2) (PGE(2)) requires intact alpha1, alpha3 and beta1 integrin subunit function, whereas epinephrine-induced hyperalgesia depends on alpha5 and beta1. PGE(2)-induced hyperalgesia is mediated by protein kinase A (PKA), while epinephrine-induced hyperalgesia is mediated by a combination of PKA, protein kinase Cepsilon (PKCepsilon) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK). We hypothesized that inflammatory mediator-induced hyperalgesia involves specific interactions between different subsets of integrin subunits and particular second messenger species. In the present study, function-blocking anti-integrin antibodies and antisense oligodeoxynucleotides were used to elucidate these interactions in rat. Hyperalgesia produced by an activator of adenylate cyclase (forskolin) depended on alpha1, alpha3 and beta1 integrins. However, hyperalgesia induced by activation of the cascade at a point farther downstream (by cAMP analog or PKA catalytic subunit) was independent of any integrins tested. In contrast, hyperalgesia induced by a specific PKCepsilon agonist depended only on alpha5 and beta1 integrins. Hyperalgesia induced by agonism of MAPK/ERK depended on all four integrin subunits tested (alpha1, alpha3, alpha5 and beta1). Finally, disruption of lipid rafts antagonized hyperalgesia induced by PGE(2) and by forskolin, but not that induced by epinephrine. Furthermore, alpha1 integrin, but not alpha5, was present in detergent-resistant membrane fractions (which retain lipid raft components). These observations suggest that integrins play a critical role in inflammatory pain by interacting with components of second messenger cascades that mediate inflammatory hyperalgesia, and that such interaction with the PGE(2)-activated pathway may be organized by lipid rafts.
Collapse
Affiliation(s)
- Olayinka A Dina
- Department of Medicine, Division of Neuroscience and Biomedical Sciences Program, NIH Pain Center (UCSF), University of California at San Francisco, Campus Box 0440, Room C-555 521, San Francisco, CA 94143-0440, USA
| | | | | | | | | | | |
Collapse
|
22
|
Allen MD, Young LS, Dawson CW. The Epstein-Barr virus-encoded LMP2A and LMP2B proteins promote epithelial cell spreading and motility. J Virol 2005; 79:1789-802. [PMID: 15650203 PMCID: PMC544088 DOI: 10.1128/jvi.79.3.1789-1802.2005] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Accepted: 09/13/2004] [Indexed: 12/26/2022] Open
Abstract
The frequent expression of latent membrane proteins LMP2A and LMP2B in Epstein Barr virus (EBV)-associated tumors suggests that these proteins play a role in EBV-induced epithelial cell growth transformation. Expression of LMP2A and LMP2B had no effect on the morphology of squamous epithelial cells in monolayer culture, but their expression was associated with an increased capacity to spread and migrate on extracellular matrix. Although the mechanisms by which LMP2A and LMP2B promote cell spreading and motility are unclear, the use of selective pharmacological inhibitors has established a role for tyrosine kinases in this phenotype but ruled out contributions of phosphatidylinositol 3-kinase, extracellular signal-regulated kinase/mitogen-activated protein kinase, and protein kinase C. The ability of LMP2B to induce a phenotype that is virtually indistinguishable from that of LMP2A suggests that regions of the LMP2 protein in addition to the cytosolic amino terminus are capable of inducing phenotypic effects in epithelial cells. Thus, rather than serving to modulate the activity of LMP2A, LMP2B may directly engage signaling pathways to influence epithelial cell behavior such as cell adhesion and motility.
Collapse
Affiliation(s)
- Michael D Allen
- Cancer Research UK Institute for Cancer Studies, University of Birmingham Medical School, Birmingham B15 2TT, United Kingdom
| | | | | |
Collapse
|
23
|
Guzeloglu A, Binelli M, Badinga L, Hansen TR, Thatcher WW. Inhibition of phorbol ester-induced PGF2alpha secretion by IFN-tau is not through regulation of protein kinase C. Prostaglandins Other Lipid Mediat 2004; 74:87-99. [PMID: 15560118 DOI: 10.1016/j.prostaglandins.2004.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Inhibitory effect of IFN-tau on phorbol ester (PdBu)-induced PGF2alpha secretion was hypothesized to be manifested by the regulation of protein kinase C (PKC) in bovine endometrial (BEND) cells. Following 12 h stimulation with PdBu, cells were unresponsive to freshly added PdBu. Pretreatment of cells with a PKC inhibitor abolished PGF2alpha secretion in response to PdBu. Therefore, PdBu induction of PGF2alpha secretion is through activation of PKC. The alpha, epsilon, iota and lambda isotypes of the PKC family were identified by Western blotting. Cells were then treated with medium alone (control), PdBu or PdBu + IFN-tau for 3 or 6 h. The PdBu-induced secretion of PGF2alpha was suppressed by IFN-tau. At 3 and 6 h, PKCalpha and PKCepsilon were detected both in the cytosolic and membrane fractions of unstimulated cells. There was a clear reduction of PKCalpha in the cytoplasm induced by PdBu and PdBu + IFN-tau at 3 and 6 h. The total abundance (cytoplasm and membrane fractions) of PKCalpha was lower in the PdBu + IFN-tau than PdBu alone. These temporal responses indicate a PKCalpha responsiveness of BEND cells to PdBu and PDBu + INF-tau with some evidence that IFN-tau causes a slight but detectable reduction in PKCalpha when added with PdBu. However, IFN-tau-induced decrease in the total abundance of PKCalpha was not enough to affect negatively the translocation of the PKCalpha to the membrane. Therefore, IFN-tau's ability to suppress secretion of PGF2alpha is unlikely due to an interference with the PdBu-induced activity of PKC.
Collapse
Affiliation(s)
- A Guzeloglu
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | | | | | | | | |
Collapse
|
24
|
Wu D, Thakore CU, Wescott GG, McCubrey JA, Terrian DM. Integrin signaling links protein kinase Cepsilon to the protein kinase B/Akt survival pathway in recurrent prostate cancer cells. Oncogene 2004; 23:8659-72. [PMID: 15467757 DOI: 10.1038/sj.onc.1207900] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Failure of hormone therapy often involves an outgrowth of protein kinase Cepsilon (PKCepsilon)-positive cells in recurrent prostate cancer. Our previous investigations have uncovered evidence of a complex signaling network operating downstream of this oncogenic protein kinase to actively advance the survival and proliferation of prostate cancer cells. In this study, we present evidence of a functional interplay among integrin receptors, PKCepsilon, and protein kinase B (PKB/Akt) in recurrent CWR-R1 prostate cancer cells. Flow cytometry and confocal microscopy provided evidence that PKCepsilon signaling promoted the assembly of matrix adhesions containing an abundance of colocalized actin filaments and beta1 integrins that exhibited an exposed activation epitope on the surface of live CWR-R1 cells. Reciprocal coimmunoprecipitations provided evidence of signaling complexes containing PKCepsilon, beta1 integrins, Src, and PKB/Akt in CWR-R1 cell cultures. An investigation into the functional significance of these interactions, and of their positive influence on beta1 integrins, demonstrated that PKCepsilon and several key components of the PKB/Akt signaling pathway remain constitutively phosphorylated/activated in adherent but not suspension cultures of PTEN-positive CWR-R1 cells. Gene transfer, antisense and pharmacological experiments provided additional support for the hypothesis that a mutually reinforcing signaling loop sustains the activation of beta1 integrins, PKCepsilon, and PKB/Akt in adherent prostate cancer cells.
Collapse
Affiliation(s)
- Daqing Wu
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | | | | | | | | |
Collapse
|
25
|
Lang W, Wang H, Ding L, Xiao L. Cooperation between PKC-alpha and PKC-epsilon in the regulation of JNK activation in human lung cancer cells. Cell Signal 2004; 16:457-67. [PMID: 14709334 DOI: 10.1016/j.cellsig.2003.09.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Phorbol esters can induce activation of two mitogen-activated protein kinase (MAPK) pathways, the extracellular signal-regulated kinase (ERK) pathway and the c-Jun N-terminal kinase (JNK) pathway. Unlike ERK activation, JNK activation by phorbol esters is somehow cell-specific. However, the mechanism(s) that contribute to the cell-specific JNK activation remain elusive. In this study, we found that phorbol 12-myristate 13-acetate (PMA) induced JNK activation only in non-small cell lung cancer (NSCLC) cells, but not in small cell lung cancer (SCLC) cells, whereas ERK activation was detected in both cell types. In NSCLC cells, PMA induced JNK activation in a time- and dose-dependent manner. JNK activation was attenuated by protein kinase C (PKC) down-regulation through prolonged pre-treatment with PMA and significantly inhibited by PKC inhibitors Gö6976 and GF109203X. Subcellular localization studies demonstrated that PMA induced translocation of PKC-alpha, -betaII, and -epsilon isoforms, but not PKC-delta, from the cytosol to the membrane. Analysis of various PKC isoforms revealed that PKC-epsilon was exclusively absent in the SCLC cell lines tested. Ectopic expression of PKC-epsilon in SCLC cells restored PMA activation of JNK signaling only in the presence of PKC-alpha, suggesting that PKC-alpha and PKC-epsilon act cooperatively in regulating JNK activation in response to PMA. Furthermore, using dominant negative mutants and pharmacological inhibitors, we define that a putative Rac1/Cdc42/PKC-alpha pathway is convergent with the PKC-epsilon/MEK1/2 pathway in terms of the activation of JNK by PMA.
Collapse
Affiliation(s)
- Wenhua Lang
- University of Florida Shands Cancer Center, Department of Anatomy and Cell Biology, College of Medicine, University of Florida, 1600 SW Archer Road, P.O. Box 100232, Gainesville, FL 32610-0232, USA
| | | | | | | |
Collapse
|
26
|
Lanni C, Mazzucchelli M, Porrello E, Govoni S, Racchi M. Differential involvement of protein kinase C alpha and epsilon in the regulated secretion of soluble amyloid precursor protein. ACTA ACUST UNITED AC 2004; 271:3068-75. [PMID: 15233804 DOI: 10.1111/j.1432-1033.2004.04240.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We investigated the differential role of protein kinase C (PKC) isoforms in the regulated proteolytic release of soluble amyloid precursor protein (sAPPalpha) in SH-SY5Y neuroblastoma cells. We used cells stably transfected with cDNAs encoding either PKCalpha or PKCepsilon in the antisense orientation, producing a reduction of the expression of PKCalpha and PKCepsilon, respectively. Reduced expression of PKCalpha and/or PKCepsilon did not modify the response of the kinase to phorbol ester stimulation, demonstrating translocation of the respective isoforms from the cytosolic fraction to specific intracellular compartments with an interesting differential localization of PKCalpha to the plasma membrane and PKCepsilon to Golgi-like structures. Reduced expression of PKCalpha significantly impaired the secretion of sAPPalpha induced by treatment with phorbol esters. Treatment of PKCalpha-deficient cells with carbachol induced a significant release of sAPPalpha. These results suggest that the involvement of PKCalpha in carbachol-induced sAPPalpha release is negligible. The response to carbachol is instead completely blocked in PKCepsilon-deficient cells suggesting the importance of PKCepsilon in coupling cholinergic receptors with APP metabolism.
Collapse
Affiliation(s)
- Cristina Lanni
- Department of Experimental and Applied Pharmacology, Centre of Excellence in Applied Biology and School of Pharmacy, University of Pavia, Viale Taramelli 14, 27100 Pavia, Italy
| | | | | | | | | |
Collapse
|
27
|
Freyer AM, Billington CK, Penn RB, Hall IP. Extracellular matrix modulates beta2-adrenergic receptor signaling in human airway smooth muscle cells. Am J Respir Cell Mol Biol 2004; 31:440-5. [PMID: 15205178 DOI: 10.1165/rcmb.2003-0241oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The airways of patients with chronic asthma commonly develop an element of fixed airway obstruction, which fails to reverse with inhaled beta2-adrenoceptor agonists. Airway remodeling refers to the structural changes of the bronchi in longstanding asthma and is characterized by increased deposition and altered ratios of extracellular matrix (ECM) proteins. We therefore assessed whether ECM proteins alter beta2-adrenoceptor signaling in human airway smooth muscle cells. We report that a fibronectin environment increases responses to beta2-adrenoceptor stimulation, whereas exposure to collagen V or laminin decreases accumulation of the second messenger cyclic AMP when compared with collagens I or IV. These differences are likely to be physiologically significant as they translate into altered phosphorylation of the downstream target VASP. The altered cAMP levels are due to differences in adenylyl cyclase activity, although expression of the relevant isoforms of enzyme appears unaltered. However, inhibition of Galphai abrogates the differences in beta2-adrenoceptor-mediated cAMP accumulation in cells exposed to different matrix factors. The difference in Galphai signaling is not due to altered Galphai expression. We conclude therefore that ECM modulates Galphai activity in human airway smooth muscle cells, and propose that these changes could contribute to the fixed airway obstruction seen in patients with chronic asthma.
Collapse
MESH Headings
- Adenylyl Cyclases/metabolism
- Airway Obstruction/drug therapy
- Airway Obstruction/metabolism
- Airway Obstruction/physiopathology
- Anti-Asthmatic Agents/pharmacology
- Asthma/drug therapy
- Asthma/metabolism
- Asthma/physiopathology
- Cell Adhesion Molecules/metabolism
- Collagen Type V/pharmacology
- Cyclic AMP/metabolism
- Extracellular Matrix/drug effects
- Extracellular Matrix/physiology
- Extracellular Matrix Proteins/metabolism
- Fibronectins/pharmacology
- GTP-Binding Protein alpha Subunit, Gi2
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Humans
- Laminin/pharmacology
- Microfilament Proteins
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phosphoproteins/metabolism
- Phosphorylation
- Proto-Oncogene Proteins/metabolism
- Receptors, Adrenergic, beta-2/drug effects
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction
- Trachea/chemistry
Collapse
Affiliation(s)
- Anette M Freyer
- Division of Therapeutics and Molecular Biology, University of Nottingham, D Floor, South Block, Queens Medical Centre, Nottingham NG7 2UH, UK.
| | | | | | | |
Collapse
|
28
|
Pinton P, Leo S, Wieckowski MR, Di Benedetto G, Rizzuto R. Long-term modulation of mitochondrial Ca2+ signals by protein kinase C isozymes. ACTA ACUST UNITED AC 2004; 165:223-32. [PMID: 15096525 PMCID: PMC2172040 DOI: 10.1083/jcb.200311061] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The modulation of Ca2+ signaling patterns during repetitive stimulations represents an important mechanism for integrating through time the inputs received by a cell. By either overexpressing the isoforms of protein kinase C (PKC) or inhibiting them with specific blockers, we investigated the role of this family of proteins in regulating the dynamic interplay of the intracellular Ca2+ pools. The effects of the different isoforms spanned from the reduction of ER Ca2+ release (PKCα) to the increase or reduction of mitochondrial Ca2+ uptake (PKCζ and PKCβ/PKCδ, respectively). This PKC-dependent regulatory mechanism underlies the process of mitochondrial Ca2+ desensitization, which in turn modulates cellular responses (e.g., insulin secretion). These results demonstrate that organelle Ca2+ homeostasis (and in particular mitochondrial processing of Ca2+ signals) is tuned through the wide molecular repertoire of intracellular Ca2+ transducers.
Collapse
Affiliation(s)
- Paolo Pinton
- Section of General Pathology, Department of Experimental and Diagnostic Medicine, Via Borsari 46, 44100 Ferrara, Italy
| | | | | | | | | |
Collapse
|
29
|
Glenn HL, Jacobson BS. Cyclooxygenase and cAMP-dependent protein kinase reorganize the actin cytoskeleton for motility in HeLa cells. CELL MOTILITY AND THE CYTOSKELETON 2003; 55:265-77. [PMID: 12845600 DOI: 10.1002/cm.10128] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The adhesion of a cell to its surrounding matrix is a key determinant in many aspects of cell behavior. Adhesion consists of distinct stages : attachment, cell spreading, motility, and/or immobilization. Interrelated signaling pathways regulate these stages, and many adhesion-related signals control the architecture of the cytoskeleton. The various cytoskeletal organizations then give rise to the specific stages of adhesion. It has been shown that arachidonic acid acts at a signaling branch point during cell attachment. Arachidonic acid is metabolized via lipoxygenase to activate actin polymerization and cell spreading. It is also metabolized by cyclooxygenase to generate small actin bundles. We have used confocal microscopy and indirect immunofluorescence to investigate the structure of these cyclooxygenase dependent actin bundles in HeLa cells. We have also employed cell migration assays and pharmacological modulation of cyclooxygenase and downstream signals. The results indicate that cyclooxygenase and PKA stimulate the formation of actin bundles that contain myosin II and associate with small focal adhesions. In addition, we demonstrate that this cytoskeletal organization correlates with increased cell motility.
Collapse
Affiliation(s)
- Honor L Glenn
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003l, USA.
| | | |
Collapse
|
30
|
Lee H, Choi BH, Suh BC, Lee SK, Kim KT. Attenuation of signal flow from P2Y6 receptor by protein kinase C-alpha in SK-N-BE(2)C human neuroblastoma cells. J Neurochem 2003; 85:1043-53. [PMID: 12716436 DOI: 10.1046/j.1471-4159.2003.01761.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Extracellular nucleotides exert a variety of biological actions through several kinds of P2 receptors in many tissues and cell types. We found that treatment with nucleotides increases intracellular Ca2+ concentration ([Ca2+]i) in SK-N-BE(2)C human neuroblastoma cells with a following order of potency: UDP > UTP > ADP >> ATP. Reverse transcription-polymerase chain reaction (RT-PCR) analysis showed that specific mRNAs coding for human P2Y1, P2Y4, and P2Y6 receptors were expressed in the cells, but Northern blot analysis revealed that P2Y6 receptors were the predominant type. Activation of protein kinase C-alpha by treatment with 1 micro m phorbol 12-myristate 13-acetate dramatically inhibited both the UDP-induced [Ca2+]i rise and inositol 1,4,5-trisphosphate (IP3) generation, whereas incubation with pertussis toxin had little effect on the responses. The UDP-induced [Ca2+]i rise and IP3 production were maintained up to 30 min after stimulation, while bradykinin-induced responses rapidly decreased to the basal level within 5 min of stimulation. Pretreatment of cells with the maximal effective concentration of UDP reduced the subsequent carbachol- or bradykinin-induced [Ca2+]i rise without inhibition of IP3 generation. Neuronal differentiation of the cells by treatment with retinoic acid for 7 days did not change the expression level of P2Y6 receptors. Taken together, the data indicate that P2Y6 receptors highly responsive to diphosphonucleotide UDP are endogenously expressed in the human neuroblastoma SK-N-BE(2)C cells and that they are involved in the modulation of other phospholipase C-coupled receptor-mediated Ca2+ mobilization by depleting the IP3-sensitive Ca2+ stores.
Collapse
Affiliation(s)
- Hyun Lee
- Department of Life Science, Division of Molecular and Life Sciences, Pohang University of Science and Technology, San 31, Hyoja-Dong, Pohang 790-784, Korea
| | | | | | | | | |
Collapse
|
31
|
Muscella A, Elia MG, Greco S, Storelli C, Marsigliante S. Activation of P2Y2 purinoceptor inhibits the activity of the Na+/K+-ATPase in HeLa cells. Cell Signal 2003; 15:115-21. [PMID: 12401526 DOI: 10.1016/s0898-6568(02)00062-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The role of ATP on regulation of the Na(+)/K(+)-ATPase activity in the human cancerous HeLa cells was investigated. HeLa cells stimulated with increasing ATP concentrations showed a dose-dependent inhibition of the Na(+)/K(+)-ATPase activity. These effects were also obtained by UTP. ATP and UTP provoked a rise in intracellular calcium concentration ([Ca(2+)](i)) persisting for at least 4 min. The inhibitor of phospholipase C, U73122, blocked the elevation of [Ca(2+)](i) provoked by ATP/UTP. The expression of mRNA for P2Y2 and P2Y6 receptors was demonstrated by RT-PCR. ATP/UTP activated PKC-alpha, -betaI and -epsilon isoforms, but not PKC-delta and -zeta. The inhibition of the Na(+)/K(+)-ATPase activity by ATP/UTP was blocked by Gö6976, a specific inhibitor of the calcium-dependent PKCs. In conclusion, our results suggest that ATP/UTP modulate Na(+)/K(+)-ATPase activity in HeLa cells through the P2Y2 purinoceptor via calcium mobilisation and activation of calcium-dependent PKCs.
Collapse
Affiliation(s)
- Antonella Muscella
- Department of Biological Sciences and Environmental Technology (DisTeBA), Laboratory of Cell Physiology, University of Lecce, Ecotekne, Via Provinciale per Monteroni, 73100 Lecce, Italy
| | | | | | | | | |
Collapse
|
32
|
Glenn HL, Jacobson BS. Arachidonic acid signaling to the cytoskeleton: the role of cyclooxygenase and cyclic AMP-dependent protein kinase in actin bundling. CELL MOTILITY AND THE CYTOSKELETON 2002; 53:239-50. [PMID: 12211105 DOI: 10.1002/cm.10072] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cell adhesion to the extracellular matrix via integrins is a primary regulatory mechanism for numerous aspects of normal cellular function. However, disruption of this interaction can result in pathology. For example, one characteristic of transformed cells is loss of adhesion dependence for viability. Adhesion also is a necessary step in tumor metastasis. It has been shown previously, in HeLa cells, that cell attachment to a gelatin-coated substrate results in the release of arachidonic acid, which is metabolized by lipoxygenase. A subsequent cascade of lipid second messengers activates protein kinase C, which triggers actin polymerization leading to cell spreading. We now demonstrate by inhibitor studies and biochemical analysis, a parallel branch of arachidonic acid signaling that reorganizes the actin cytoskeleton into small bundles. This branch of the pathway is initiated by cyclooxygenase, which generates prostaglandins and causes the downstream activation of cyclic AMP-dependent protein kinase. This work elucidates a system of interacting signals in which arachidonic acid functions at a branch point in cytoskeletal signaling. The lipoxygenase branch provides polymerized actin; these actin filaments act as a substrate for the cylooxygenase branch to generate actin bundles.
Collapse
Affiliation(s)
- Honor L Glenn
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, 01003, USA
| | | |
Collapse
|
33
|
Ai H, Mills DK, Jonathan AS, Jones SA. Gelatin-glutaraldehyde cross-linking on silicone rubber to increase endothelial cell adhesion and growth. In Vitro Cell Dev Biol Anim 2002; 38:487-92. [PMID: 12703975 DOI: 10.1290/1071-2690(2002)038<0487:gcosrt>2.0.co;2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Silicone is a biomaterial that is widely used in many areas because of its high optical clarity, its durability, and the ease with which it can be cast. However, these advantages are counterbalanced by strong hydrophobicity. Gelatin cross-linking has been used as a hydrophilic coating on many biomaterials but not on silicone rubber. In this study, two gelatin glutaraldehyde (GA) cross-linking methods were used to coat a hydrophilic membrane on silicone rubber. In method I, gelatin and GA were mixed in three different proportions (64:1, 128:1, and 256:1) before coating. In method II, a newly formed 5% gelatin membrane was cross-linked with a 2.5% GA solution. All coatings were hydrophilic, as determined from the measurement of contact angle for a drop of water on the surface. Bovine coronary arterial endothelial cells were shown to grow well on the surface modified by method II at 72 h. In method I, the cells grew well for gelatin-GA proportions of 64:1 and 128:1 at 72 h. No cell attachment on untreated silicone rubber was observed by the third d of seeding. The results indicated that both methods of gelatin-GA cross-linking provided a hydrophilic surface on silicone for endothelial cell adhesion and growth in vitro.
Collapse
Affiliation(s)
- Hua Ai
- Department of Biomedical Engineering & School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana 71272, USA
| | | | | | | |
Collapse
|
34
|
VanWinkle WB, Snuggs MB, De Hostos EL, Buja LM, Woods A, Couchman JR. Localization of the transmembrane proteoglycan syndecan-4 and its regulatory kinases in costameres of rat cardiomyocytes: a deconvolution microscopic study. THE ANATOMICAL RECORD 2002; 268:38-46. [PMID: 12209563 DOI: 10.1002/ar.10130] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Syndecan-4 (syn-4), a transmembrane heparan sulfate-containing proteoglycan, is unique among the four members of the syndecan family in its specific cellular localization to complex cytoskeletal adhesion sites, i.e., focal adhesions. During early phenotypic redifferentiation of neonatal cardiomyocytes in culture, immunolocalization reveals syn-4 to be heavily concentrated in the perinuclear endoplasmic reticulum-Golgi region, with little found at the peripheral regions. Subsequently, syn-4 becomes localized to a cytoskeletal adhesion complex unique to striated muscle, the costamere. Soon after redifferentiation of myofibrils in cultured neonatal cardiomyocytes, syn-4 is present only in costameres, not in focal adhesions. In cultured adult cardiomyocytes, it is present in both costameres and focal adhesions-the latter in two distinct regions of the spread cardiomyocytes, reflecting localization with two types of actin-containing filaments. The fact that syn-4 is observed early in the costameric regions, as opposed to later in the focal adhesions, suggests that it may play an initial role in early adhesion/signal transduction mechanisms in close proximity to the contractile apparatus, as well as in transmission of contractile force to the collagenous extracellular matrix (ECM) which surrounds the cardiac myofibers in situ. With respect to possible regulatory mechanisms of syn-4, we localized syn-4 with both the epsilon isoform of protein kinase C and the tyrosine kinase pp60(csrc) in costameric regions. These findings suggest that syn-4 may not only play a role in cellular adhesion and contractile force transmission, it may also, through ser, thr, and tyr phosphorylation, be part of an interactive signal transduction mechanism in myocardial functioning via these adhesive cytoskeletal complexes.
Collapse
Affiliation(s)
- W Barry VanWinkle
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, Houston, Texas 77030, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Ivaska J, Whelan RD, Watson R, Parker PJ. PKC epsilon controls the traffic of beta1 integrins in motile cells. EMBO J 2002; 21:3608-19. [PMID: 12110574 PMCID: PMC126116 DOI: 10.1093/emboj/cdf371] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Protein kinase C (PKC) has been implicated in beta 1 integrin-mediated cell migration. Expression of the novel PKC isoform, PKC epsilon, in PKC epsilon(-/-) cells is shown here to stimulate directional migration of cells towards beta 1 integrin substrates in a manner dependent on PKC catalytic activity. On PKC inhibition, integrin beta 1 and PKC epsilon become reversibly trapped in a tetraspanin (CD81)-positive intracellular compartment, correlating with reduced haptotaxis. Immunofluorescence and pulse labelling studies indicate that this is a previously uncharacterized recycling compartment trapped by inhibition of PKC. Electron microscopy demonstrated the co-localization of PKC epsilon and integrin beta 1 on the vesicular membranes. Finally, using a reconstituted in vitro system, the dissociation of PKC epsilon from these vesicles is shown to be dependent on both the presence of cytosolic components and energy, and on PKC catalytic activity. The evidence presented indicates that PKC epsilon controls an internal traffic step that under uninhibited conditions permits the recycling of beta 1 integrin, contributing to cell motility.
Collapse
Affiliation(s)
| | | | - Rose Watson
- Protein Phosphorylation Laboratory and
Electron Microscopy Unit, Cancer Research UK London Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3PX, UK Corresponding author e-mail:
| | - Peter J. Parker
- Protein Phosphorylation Laboratory and
Electron Microscopy Unit, Cancer Research UK London Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3PX, UK Corresponding author e-mail:
| |
Collapse
|
36
|
Stapleton G, Malliri A, Ozanne BW. Downregulated AP-1 activity is associated with inhibition of Protein-Kinase-C-dependent CD44 and ezrin localisation and upregulation of PKC theta in A431 cells. J Cell Sci 2002; 115:2713-24. [PMID: 12077362 DOI: 10.1242/jcs.115.13.2713] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Progression to an invasive, metastatic tumour requires the coordinated expression and function of a number of gene products, as well as their regulation in the context of invasion. The transcription factor AP-1 regulates expression of many of those genes necessary for implementation of the invasion programme. Two such gene products, CD44 and ezrin, are both upregulated in fibroblasts transformed by v-fos and are commonly implicated in cell motility and invasion. Here we report that CD44 and ezrin colocalise to membrane ruffles and microvilli of A431 cells after treatment with EGF. However, A431 cells expressing dominant-negative c-Jun (TAM67), and which as a consequence fail to invade in response to EGF, also fail to correctly localise CD44 and ezrin. CD44 and ezrin are both substrates for Protein Kinase C, and we show that their EGF-dependent colocalisation requires Protein Kinase C activity. Associated with TAM67 expression and disrupted CD44 and ezrin colocalisation is the increased expression and activation of the novel PKC theta isoform. Expression of PKC theta in A431 cells results in the inhibition of cell motility and disrupted localisation of CD44 and ezrin. We propose that AP-1 regulates the integrity of Protein Kinase C signalling and identifies PKC theta as a potential suppressor of the invasion programme.
Collapse
Affiliation(s)
- Genevieve Stapleton
- Cancer Research UK Beatson Laboratories, Garscube Estate, Switchback Road, Glasgow, G61 1BD Scotland.
| | | | | |
Collapse
|
37
|
Disatnik MH, Boutet SC, Lee CH, Mochly-Rosen D, Rando TA. Sequential activation of individual PKC isozymes in integrin-mediated muscle cell spreading: a role for MARCKS in an integrin signaling pathway. J Cell Sci 2002; 115:2151-63. [PMID: 11973356 DOI: 10.1242/jcs.115.10.2151] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
To understand how muscle cell spreading and survival are mediated by integrins, we studied the signaling events initiated by the attachment of muscle cells to fibronectin (FN). We have previously demonstrated that muscle cell spreading on FN is mediated by alpha5beta1 integrin, is associated with rapid phosphorylation of focal adhesion kinase and is dependent on activation of protein kinase C (PKC). Here we investigated the role of individual PKC isozymes in these cellular processes. We show that alpha, delta and epsilonPKC are expressed in muscle cells and are activated upon integrin engagement with different kinetics - epsilonPKC was activated early, whereas alpha and deltaPKC were activated later. Using isozyme-specific inhibitors, we found that the activation of epsilonPKC was necessary for cell attachment to FN. However, using isozyme-specific activators, we found that activation of each of three isozymes was sufficient to promote the spreading of alpha5-integrin-deficient cells on FN. To investigate further the mechanism by which integrin signaling and PKC activation mediate cell spreading, we studied the effects of these processes on MARCKS, a substrate of PKC and a protein known to regulate actin dynamics. We found that MARCKS was localized to focal adhesion sites soon after cell adhesion and that MARCKS translocated from the membrane to the cytosol during the process of cell spreading. This translocation correlated with different phases of PKC activation and with reorganization of the actin cytoskeleton. Using MARCKS-antisense cDNA, we show that alpha5-expressing cells in which MARCKS expression is inhibited fail to spread on FN, providing evidence for the crucial role of MARCKS in muscle cell spreading. Together, the data suggest a model in which early activation of epsilonPKC is necessary for cell attachment; the later activation of alpha or deltaPKC may be necessary for the progression from attachment to spreading. The mechanism of PKC-mediated cell spreading may be via the phosphorylation of signaling proteins, such as MARCKS, that are involved in the reorganization of the actin cytoskeleton.
Collapse
Affiliation(s)
- Marie-Hélène Disatnik
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, CA 94305, USA
| | | | | | | | | |
Collapse
|
38
|
Cenni V, Döppler H, Sonnenburg ED, Maraldi N, Newton AC, Toker A. Regulation of novel protein kinase C epsilon by phosphorylation. Biochem J 2002; 363:537-45. [PMID: 11964154 PMCID: PMC1222506 DOI: 10.1042/0264-6021:3630537] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The activity and intracellular localization of protein kinase C (PKC) family members are controlled by phosphorylation at three highly conserved sites in the catalytic kinase domain. In the case of the novel PKCepsilon isoform, these are Thr(566) in the activation loop, Thr(710) in the turn motif and Ser(729) in the C-terminal hydrophobic motif. In the present study, we analysed the contribution of the phosphoinositide-dependent kinase 1 (PDK-1) and PKCepsilon kinase activity in controlling the phosphorylation of Thr(566) and Ser(729). In NIH 3T3 fibroblasts, PKCepsilon migrated as a single band, and stimulation with platelet-derived growth factor resulted in the appearance of a second band with a slower electrophoretic mobility, concomitant with an increase in phosphorylation of Thr(566) and Ser(729). Cells transfected with an active PDK-1 allele also resulted in increased PKCepsilon Thr(566) and Ser(729) phosphorylation, whereas an active myristoylated PKCepsilon mutant was constitutively phosphorylated at these sites. Protein kinase-inactive mutants of PKCepsilon were not phosphorylated at Ser(729) in cells, and phosphorylation of this site leads to dephosphorylation of the activation-loop Thr(566), an effect which can be reversed with either okadaic acid or co-transfection with active PDK-1. In vitro, PDK-1 catalysed the phosphorylation of purified PKCepsilon in the presence of mixed micelles containing either diacylglycerol or PtdIns(3,4,5)P(3), concomitant with an increase in Ser(729) phosphorylation. These studies reveal that the mechanism of phosphorylation of a novel PKC is the same as that for conventional PKCs: PDK-1 phosphorylation of the activation loop triggers autophosphorylation of the hydrophobic motif. However, the regulation of this phosphorylation is different for novel and conventional PKCs. Specifically, the phosphorylation of novel PKCs is regulated rather than constitutive.
Collapse
Affiliation(s)
- Vittoria Cenni
- Boston Biomedical Research Institute, Watertown, MA 02472, USA
| | | | | | | | | | | |
Collapse
|
39
|
Ridge KM, Dada L, Lecuona E, Bertorello AM, Katz AI, Mochly-Rosen D, Sznajder JI. Dopamine-induced exocytosis of Na,K-ATPase is dependent on activation of protein kinase C-epsilon and -delta. Mol Biol Cell 2002; 13:1381-9. [PMID: 11950946 PMCID: PMC102276 DOI: 10.1091/mbc.01-07-0323] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The purpose of this study was to define mechanisms by which dopamine (DA) regulates the Na,K-ATPase in alveolar epithelial type 2 (AT2) cells. The Na,K-ATPase activity increased by twofold in cells incubated with either 1 microM DA or a dopaminergic D(1) agonist, fenoldopam, but not with the dopaminergic D(2) agonist quinpirole. The increase in activity paralleled an increase in Na,K-ATPase alpha1 and beta1 protein abundance in the basolateral membrane (BLM) of AT2 cells. This increase in protein abundance was mediated by the exocytosis of Na,K-pumps from late endosomal compartments into the BLM. Down-regulation of diacylglycerol-sensitive types of protein kinase C (PKC) by pretreatment with phorbol 12-myristate 13-acetate or inhibition with bisindolylmaleimide prevented the DA-mediated increase in Na,K-ATPase activity and exocytosis of Na,K-pumps to the BLM. Preincubation of AT2 cells with either 2-[1-(3-dimethylaminopropyl)-5-methoxyindol-3-yl]-3-(1H-indol-3-yl)maleimide (Gö6983), a selective inhibitor of PKC-delta, or isozyme-specific inhibitor peptides for PKC-delta or PKC-epsilon inhibited the DA-mediated increase in Na,K-ATPase. PKC-delta and PKC-epsilon, but not PKC-alpha or -beta, translocated from the cytosol to the membrane fraction after exposure to DA. PKC-delta- and PKC-epsilon-specific peptide agonists increased Na,K-ATPase protein abundance in the BLM. Accordingly, dopamine increased Na,K-ATPase activity in alveolar epithelial cells through the exocytosis of Na,K-pumps from late endosomes into the basolateral membrane in a mechanism-dependent activation of the novel protein kinase C isozymes PKC-delta and PKC-epsilon.
Collapse
Affiliation(s)
- Karen M Ridge
- Pulmonary and Critical Care Medicine, Northwestern University Medical School, Chicago, IL 60611, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Ziegler WH, Tigges U, Zieseniss A, Jockusch BM. A lipid-regulated docking site on vinculin for protein kinase C. J Biol Chem 2002; 277:7396-404. [PMID: 11741957 DOI: 10.1074/jbc.m110008200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
During cell spreading, binding of actin-organizing proteins to acidic phospholipids and phosphorylation are important for localization and activity of these proteins at nascent cell-matrix adhesion sites. Here, we report on a transient interaction between the lipid-dependent protein kinase Calpha and vinculin, an early component of these sites, during spreading of HeLa cells on collagen. In vitro binding of protein kinase Calpha to vinculin tail was found dependent on free calcium and acidic phospholipids but independent of a functional kinase domain. The interaction was enhanced by conditions that favor the oligomerization of vinculin. Phosphorylation by protein kinase Calpha reached 1.5 mol of phosphate/mol of vinculin tail and required the C-terminal hydrophobic hairpin, a putative phosphatidylinositol 4,5-bisphosphate-binding site. Mass spectroscopy of peptides derived from in vitro phosphorylated vinculin tail identified phosphorylation of serines 1033 and 1045. Inhibition of C-terminal phospholipid binding at the vinculin tail by mutagenesis or deletion reduced the rate of phosphorylation to < or =50%. We suggest a possible mechanism whereby phospholipid-regulated conformational changes in vinculin may lead to exposure of a docking site for protein kinase Calpha and subsequent phosphorylation of vinculin and/or vinculin interaction partners, thereby affecting the formation of cell adhesion complexes.
Collapse
Affiliation(s)
- Wolfgang H Ziegler
- Department of Cell Biology, Zoological Institute, Technical University of Braunschweig, D-38092 Braunschweig, Germany
| | | | | | | |
Collapse
|
41
|
Saitoh N, Hori T, Takahashi T. Activation of the epsilon isoform of protein kinase C in the mammalian nerve terminal. Proc Natl Acad Sci U S A 2001; 98:14017-21. [PMID: 11717460 PMCID: PMC61159 DOI: 10.1073/pnas.241333598] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Activation of protein kinase C (PKC) by phorbol ester facilitates hormonal secretion and transmitter release, and phorbol ester-induced synaptic potentiation (PESP) is a model for presynaptic facilitation. A variety of PKC isoforms are expressed in the central nervous system, but the isoform involved in the PESP has not been identified. To address this question, we have applied immunocytochemical and electrophysiological techniques to the calyx of Held synapse in the medial nucleus of the trapezoid body (MNTB) of rat auditory brainstem. Western blot analysis indicated that both the Ca(2+)-dependent "conventional" PKC and Ca(2+)-independent "novel" PKC isoforms are expressed in the MNTB. Denervation of afferent fibers followed by organotypic culture, however, selectively decreased "novel" epsilon PKC isoform expressed in this region. The afferent calyx terminal was clearly labeled with the epsilon PKC immunofluorescence. On stimulation with phorbol ester, presynaptic epsilon PKC underwent autophosphorylation and unidirectional translocation toward the synaptic side. Chelating presynaptic Ca(2+), by using membrane permeable EGTA analogue or high concentration of EGTA directly loaded into calyceal terminals, had only a minor attenuating effect on the PESP. We conclude that the Ca(2+)-independent epsilon PKC isoform mediates the PESP at this mammalian central nervous system synapse.
Collapse
Affiliation(s)
- N Saitoh
- Department of Neurophysiology, University of Tokyo, Graduate School of Medicine, Tokyo 113-0033, Japan
| | | | | |
Collapse
|
42
|
Suh BC, Lee H, Jun DJ, Chun JS, Lee JH, Kim KT. Inhibition of H2 histamine receptor-mediated cation channel opening by protein kinase C in human promyelocytic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:1663-71. [PMID: 11466390 DOI: 10.4049/jimmunol.167.3.1663] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Histamine, through H(2) receptors, triggers a prominent rise in intracellular free Ca(2+) concentration ([Ca(2+)](i)) in addition to an elevation of cAMP level in HL-60 promyelocytes. Here we show that the histamine-induced [Ca(2+)](i) rise was due to influx of Ca(2+) from the extracellular space, probably through nonselective cation channels, as incubation of the cells with SKF 96365 abolished the histamine-induced [Ca(2+)](i) rise, Na(+) influx, and membrane depolarization. The Ca(2+) influx was specifically inhibited by pretreatment of the cells with PMA or extracellular ATP with 50% inhibitory concentrations of 0.12 +/- 0.03 nM and 185 +/- 17 microM, respectively. Western blot analysis of protein kinase C (PKC) isoforms revealed that PMA (< or =1 nM) and ATP (300 microM) caused selective translocation of PKC-delta to the particulate/membrane fraction. Costimulation of the cells with histamine and SKF 96365 partially reduced histamine-induced granulocytic differentiation, which was evaluated by looking at the extent of fMet-Leu-Phe-induced [Ca(2+)](i) rise and superoxide generation. In conclusion, nonselective cation channels are opened by stimulation of the H(2) receptor, and the channels are at least in part involved in the induction of histamine-mediated differentiation processes. Both effects of histamine were selectively inhibited probably by the delta isoform of PKC in HL-60 cells.
Collapse
Affiliation(s)
- B C Suh
- Department of Life Science, Division of Molecular and Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
| | | | | | | | | | | |
Collapse
|
43
|
Stockton RA, Jacobson BS. Modulation of cell-substrate adhesion by arachidonic acid: lipoxygenase regulates cell spreading and ERK1/2-inducible cyclooxygenase regulates cell migration in NIH-3T3 fibroblasts. Mol Biol Cell 2001; 12:1937-56. [PMID: 11451994 PMCID: PMC55641 DOI: 10.1091/mbc.12.7.1937] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adhesion of cells to an extracellular matrix is characterized by several discrete morphological and functional stages beginning with cell-substrate attachment, followed by cell spreading, migration, and immobilization. We find that although arachidonic acid release is rate-limiting in the overall process of adhesion, its oxidation by lipoxygenase and cyclooxygenases regulates, respectively, the cell spreading and cell migration stages. During the adhesion of NIH-3T3 cells to fibronectin, two functionally and kinetically distinct phases of arachidonic acid release take place. An initial transient arachidonate release occurs during cell attachment to fibronectin, and is sufficient to signal the cell spreading stage after its oxidation by 5-lipoxygenase to leukotrienes. A later sustained arachidonate release occurs during and after spreading, and signals the subsequent migration stage through its oxidation to prostaglandins by newly synthesized cyclooxygenase-2. In signaling migration, constitutively expressed cyclooxygenase-1 appears to contribute approximately 25% of prostaglandins synthesized compared with the inducible cyclooxygenase-2. Both the second sustained arachidonate release, and cyclooxygenase-2 protein induction and synthesis, appear to be regulated by the mitogen-activated protein kinase extracellular signal-regulated kinase (ERK)1/2. The initial cell attachment-induced transient arachidonic acid release that signals spreading through lipoxygenase oxidation is not sensitive to ERK1/2 inhibition by PD98059, whereas PD98059 produces both a reduction in the larger second arachidonate release and a blockade of induced cyclooxygenase-2 protein expression with concomitant reduction of prostaglandin synthesis. The second arachidonate release, and cyclooxygenase-2 expression and activity, both appear to be required for cell migration but not for the preceding stages of attachment and spreading. These data suggest a bifurcation in the arachidonic acid adhesion-signaling pathway, wherein lipoxygenase oxidation generates leukotriene metabolites regulating the spreading stage of cell adhesion, whereas ERK 1/2-induced cyclooxygenase synthesis results in oxidation of a later release, generating prostaglandin metabolites regulating the later migration stage.
Collapse
Affiliation(s)
- R A Stockton
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | |
Collapse
|
44
|
Shi M, Dennis K, Peschon JJ, Chandrasekaran R, Mikecz K. Antibody-induced shedding of CD44 from adherent cells is linked to the assembly of the cytoskeleton. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:123-31. [PMID: 11418640 DOI: 10.4049/jimmunol.167.1.123] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
CD44 is a widely expressed integral membrane glycoprotein that serves as a specific adhesion receptor for the extracellular matrix glycosaminoglycan hyaluronan. CD44 participates in a variety of physiological and pathological processes through its role in cell adhesion. Under appropriate conditions, the ectodomain of CD44 is proteolytically removed from the cell surface. In this study we show that excessive CD44 shedding can be induced in mouse fibroblasts and monocytes upon exposure of these cells to a CD44-specific Ab immobilized on plastic, whereas treatment with phorbol ester induces significantly enhanced CD44 release from the monocytes only. CD44 shedding proceeds normally in fibroblasts and monocytes deficient in TNF-alpha converting enzyme (TACE), a sheddase involved in the processing of several substrates. Conversely, activation of the CD44 protease has no effect on the release of TNF-alpha from TACE-expressing cells, although the same metalloprotease inhibitor effectively blocks both TACE and the CD44 sheddase. Concomitant with anti-CD44 Ab- or phorbol ester-induced CD44 shedding, dramatic changes are observed in cell morphology and the structure of the actin cytoskeleton. Disruption of actin assembly with cytochalasin reduces CD44 shedding, but not the release of TNF-alpha. Moreover, pharmacological activation of Rho family GTPases Rac1 and Cdc42, which regulate actin filament assembly into distinct cytoskeletal structures, has a profound effect on CD44 release. We conclude that the CD44 sheddase and TACE are distinct enzymes, and that Ab- and phorbol ester-enhanced cleavage of CD44 is controlled in a cell type-dependent fashion by Rho GTPases through the cytoskeleton.
Collapse
Affiliation(s)
- M Shi
- Departments of. Biochemistry and Orthopedic Surgery, Section of Biochemistry and Molecular Biology, Rush University at Rush-Presbyterian-St. Luke's Medical Center, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
45
|
Berrier AL, Mastrangelo AM, Downward J, Ginsberg M, LaFlamme SE. Activated R-ras, Rac1, PI 3-kinase and PKCepsilon can each restore cell spreading inhibited by isolated integrin beta1 cytoplasmic domains. J Cell Biol 2000; 151:1549-60. [PMID: 11134082 PMCID: PMC2150687 DOI: 10.1083/jcb.151.7.1549] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2000] [Accepted: 11/06/2000] [Indexed: 11/30/2022] Open
Abstract
Attachment of many cell types to extracellular matrix proteins triggers cell spreading, a process that strengthens cell adhesion and is a prerequisite for many adhesion-dependent processes including cell migration, survival, and proliferation. Cell spreading requires integrins with intact beta cytoplasmic domains, presumably to connect integrins with the actin cytoskeleton and to activate signaling pathways that promote cell spreading. Several signaling proteins are known to regulate cell spreading, including R-Ras, PI 3-kinase, PKCepsilon and Rac1; however, it is not known whether they do so through a mechanism involving integrin beta cytoplasmic domains. To study the mechanisms whereby cell spreading is regulated by integrin beta cytoplasmic domains, we inhibited cell spreading on collagen I or fibrinogen by expressing tac-beta1, a dominant-negative inhibitor of integrin function, and examined whether cell spreading could be restored by the coexpression of either V38R-Ras, p110alpha-CAAX, myr-PKCepsilon, or L61Rac1. Each of these activated signaling proteins was able to restore cell spreading as assayed by an increase in the area of cells expressing tac-beta1. R-Ras and Rac1 rescued cell spreading in a GTP-dependent manner, whereas PKCstraightepsilon required an intact kinase domain. Importantly, each of these signaling proteins required intact beta cytoplasmic domains on the integrins mediating adhesion in order to restore cell spreading. In addition, the rescue of cell spreading by V38R-Ras was inhibited by LY294002, suggesting that PI 3-kinase activity is required for V38R-Ras to restore cell spreading. In contrast, L61Rac1 and myr-PKCstraightepsilon each increased cell spreading independent of PI 3-kinase activity. Additionally, the dominant-negative mutant of Rac1, N17Rac1, abrogated cell spreading and inhibited the ability of p110alpha-CAAX and myr-PKCstraightepsilon to increase cell spreading. These studies suggest that R-Ras, PI 3-kinase, Rac1 and PKCepsilon require the function of integrin beta cytoplasmic domains to regulate cell spreading and that Rac1 is downstream of PI 3-kinase and PKCepsilon in a pathway involving integrin beta cytoplasmic domain function in cell spreading.
Collapse
Affiliation(s)
- Allison L. Berrier
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208
| | - Anthony M. Mastrangelo
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208
| | - Julian Downward
- Imperial Cancer Research Fund, London WC2A 3PX, United Kingdom
| | - Mark Ginsberg
- Department of Vascular Biology, The Scripps Research Institute, La Jolla, California 92037
| | - Susan E. LaFlamme
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208
| |
Collapse
|
46
|
Khatib AM, Nip J, Fallavollita L, Lehmann M, Jensen G, Brodt P. Regulation of urokinase plasminogen activator/plasmin- mediated invasion of melanoma cells by the integrin vitronectin receptor αVβ3. Int J Cancer 2000. [DOI: 10.1002/1097-0215(200002)9999:9999<::aid-ijc1055>3.0.co;2-n] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
47
|
Kang Y, Park JS, Kim SH, Shin YJ, Kim W, Joo HJ, Chun JS, Kim HJ, Ha MJ. Overexpression of protein kinase C delta represses expression of proliferin in NIH3T3 cells that regulates cell proliferation. MOLECULAR CELL BIOLOGY RESEARCH COMMUNICATIONS : MCBRC 2000; 4:181-7. [PMID: 11281734 DOI: 10.1006/mcbr.2001.0276] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Protein kinase C (PKC) delta is known to inhibit proliferation of many cell types. In this study we found that overexpression of PKCdelta reduced proliferation of NIH3T3 cells. To identify specific genes regulated by PKCdelta in regulation of cell proliferation, we used differential display-polymerase chain reaction in PKCdelta-overexpressing NIH3T3 cells and found that the expression of proliferin, a secreted protein known to stimulate cell proliferation, was significantly repressed. Transient transfection study indicated that the repression of proliferin expression was inversely proportional to the expression levels of PKCdelta. Addition of an anti-proliferin antibody to culture medium to neutralize the secreted proliferin decreased cell proliferation in a dose-dependent manner. Our results, therefore, suggest that overexpression of PKCdelta induces transcriptional repression of proliferin, thereby resulting in inhibition of cell proliferation.
Collapse
Affiliation(s)
- Y Kang
- Institute for Medical Sciences, Ajou University School of Medicine, Suwon 442-721, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Oh CD, Chang SH, Yoon YM, Lee SJ, Lee YS, Kang SS, Chun JS. Opposing role of mitogen-activated protein kinase subtypes, erk-1/2 and p38, in the regulation of chondrogenesis of mesenchymes. J Biol Chem 2000; 275:5613-9. [PMID: 10681543 DOI: 10.1074/jbc.275.8.5613] [Citation(s) in RCA: 182] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The present studies were performed to determine subtype-specific roles of mitogen-activated protein kinase in chondrogenesis. Erk-1/2 activities, downstream of protein kinase C, decreased as chondrogenesis proceeded, whereas p38 activities, independent of protein kinase C, continuously increased during chondrogenesis. Inhibition of Erk-1/2 with PD98059 enhanced chondrogenesis up to 1. 7-fold, whereas inhibition of p38 with SB203580 reduced it to about 30% of the control level. Inhibition of Erk-1/2 or p38 did not affect precartilage condensation. However, cartilage nodule formation was significantly blocked by the inhibition of p38, whereas Erk-1/2 inhibition did not affect it. Modulation of chondrogenesis by the inhibition of Erk-1/2 and p38 was accompanied by altered expression of adhesion molecules in an opposite way. Expression of N-cadherin was reduced as chondrogenesis proceeded. Inhibition of p38 caused sustained expression of N-cadherin, whereas Erk-1/2 inhibition accelerated the reduction of N-cadherin expression. Expression of integrin alpha5beta1 and fibronectin were found to transiently increase during chondrogenesis. Inhibition of p38 caused continuous increase of expression of these molecules, whereas Erk-1/2 inhibition accelerated the decrease of expression of these molecules at a later period of chondrogenesis. Because temporal expression of these adhesion molecules regulates chondrogenesis, the above results indicate that Erk-1/2 and p38 conversely regulate chondrogenesis at post-precartilage condensation stages by modulating expression of adhesion molecules.
Collapse
Affiliation(s)
- C D Oh
- Department of Biology, Kyungpook National University, Pook-Gu, Taegu 702-701, Korea
| | | | | | | | | | | | | |
Collapse
|
49
|
Shark cartilage extract interferes with cell adhesion and induces reorganization of focal adhesions in cultured endothelial cells. J Cell Biochem 2000. [DOI: 10.1002/1097-4644(20000901)78:3<417::aid-jcb7>3.0.co;2-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
50
|
Defilippi P, Olivo C, Venturino M, Dolce L, Silengo L, Tarone G. Actin cytoskeleton organization in response to integrin-mediated adhesion. Microsc Res Tech 1999; 47:67-78. [PMID: 10506763 DOI: 10.1002/(sici)1097-0029(19991001)47:1<67::aid-jemt7>3.0.co;2-p] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cell matrix adhesion regulates actin cytoskeleton organization through distinct steps, from formation of filopodia and lamellipodia in the early phases of cell adhesion to organization of focal adhesions and stress fibers in fully adherent cells. In this review, we follow the events induced by integrin-mediated adhesion, such as activation of GTPases Cdc42 and Rac and their effectors and their role in actin polymerization leading to formation of lamellipodia and filopodia and cell spreading. We also show that actin stress fiber and focal adhesion formation following adhesion requires cooperation between integrin-mediated signaling and additional stimuli, including activation of PKC, Rho GTPases, and PTKs such as p125Fak and Src.
Collapse
Affiliation(s)
- P Defilippi
- Dipartimento di Genetica, Biologia e Biochimica, Università di Torino, Torino, Italy.
| | | | | | | | | | | |
Collapse
|