1
|
Knaup I, Symmank J, Bastian A, Neuss S, Pufe T, Jacobs C, Wolf M. Impact of FGF1 on human periodontal ligament fibroblast growth, osteogenic differentiation and inflammatory reaction in vitro. J Orofac Orthop 2021; 83:42-55. [PMID: 34874457 DOI: 10.1007/s00056-021-00363-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE To investigate in vitro the impact of fibroblast growth factor 1 (FGF1) in comparison to ascorbic acid (AscA) on human periodontal ligament fibroblast (HPdLF) growth, their osteogenic differentiation, and modulation of their inflammatory reaction to mechanical stress. METHODS The influence of different concentrations of FGF1 (12.5-200 ng/mL) on growth and proliferation of HPdLF cells was analyzed over 20 days by counting cell numbers and the percentage of Ki67-positive cells. Quantitative expression analysis of genes encoding the osteogenic markers alkaline phosphatase (ALPL), Runt-related transcription factor 2 (RUNX2), osteocalcin (OCN), and osteopontin (OSP), as well as the fibroblast markers vimentin (VIM) and fibroblast-specific protein 1 (FSP1), was performed after 2 and 20 days of cultivation. Metabolic activity was determined by MTT assay. For comparison with AscA, 50 ng/mL FGF1 was used for stimulation for 2 and 20 days. Cell number, percentage of Ki67-positive cells, and expression of osteoblast- and fibroblast-specific genes were examined. Alkaline phosphatase activity was visualized by NBT/BCIP and calcium deposits were stained with alizarin red. Cytokine (IL‑6, IL‑8, COX2/PGE2) expression and secretion were analyzed by qPCR and ELISA in 6 h mechanically compressed HPdLF cultured for 2 days with FGF1 or ascorbic acid. RESULTS Higher concentrations of FGF1 promoted cell proliferation upon short-term stimulation, whereas prolonged treatment induced the expression of osteogenic markers even with low concentrations. AscA promotes cell growth more markedly than FGF1 in short-term cultures, whereas FGF1 induced osteogenic cell fate more strongly in long-term culture. Both factors induced an increased inflammatory response of HPdLF to mechanical compression. CONCLUSION Our data suggest that FGF1 promotes an osteogenic phenotype of HPdLF and limits inflammatory response to mechanical forces compared to AscA.
Collapse
Affiliation(s)
- Isabel Knaup
- Department of Orthodontics, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Judit Symmank
- Department of Orthodontics, Jena University Hospital, Jena, Germany
| | - Asisa Bastian
- Department of Orthodontics, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Sabine Neuss
- Helmholtz Institute for Biomedical Engineering, BioInterface Group, RWTH Aachen University, Aachen, Germany
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University Hospital, Wendlingweg 2, 52074, Aachen, Germany
| | - Collin Jacobs
- Department of Orthodontics, Jena University Hospital, Jena, Germany
| | - Michael Wolf
- Department of Orthodontics, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| |
Collapse
|
2
|
Sluzalska KD, Slawski J, Sochacka M, Lampart A, Otlewski J, Zakrzewska M. Intracellular partners of fibroblast growth factors 1 and 2 - implications for functions. Cytokine Growth Factor Rev 2020; 57:93-111. [PMID: 32475760 DOI: 10.1016/j.cytogfr.2020.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Fibroblast growth factors 1 and 2 (FGF1 and FGF2) are mainly considered as ligands of surface receptors through which they regulate a broad spectrum of biological processes. They are secreted in non-canonical way and, unlike other growth factors, they are able to translocate from the endosome to the cell interior. These unique features, as well as the role of the intracellular pool of FGF1 and FGF2, are far from being fully understood. An increasing number of reports address this problem, focusing on the intracellular interactions of FGF1 and 2. Here, we summarize the current state of knowledge of the FGF1 and FGF2 binding partners inside the cell and the possible role of these interactions. The partner proteins are grouped according to their function, including proteins involved in secretion, cell signaling, nucleocytoplasmic transport, binding and processing of nucleic acids, ATP binding, and cytoskeleton assembly. An in-depth analysis of the network of these binding partners could indicate novel, non-classical functions of FGF1 and FGF2 and uncover an additional level of a fine control of the well-known FGF-regulated cellular processes.
Collapse
Affiliation(s)
- Katarzyna Dominika Sluzalska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jakub Slawski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Martyna Sochacka
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Agata Lampart
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
3
|
Kostas M, Lampart A, Bober J, Wiedlocha A, Tomala J, Krowarsch D, Otlewski J, Zakrzewska M. Translocation of Exogenous FGF1 and FGF2 Protects the Cell against Apoptosis Independently of Receptor Activation. J Mol Biol 2018; 430:4087-4101. [DOI: 10.1016/j.jmb.2018.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 01/16/2023]
|
4
|
Sampayo RG, Toscani AM, Rubashkin MG, Thi K, Masullo LA, Violi IL, Lakins JN, Cáceres A, Hines WC, Coluccio Leskow F, Stefani FD, Chialvo DR, Bissell MJ, Weaver VM, Simian M. Fibronectin rescues estrogen receptor α from lysosomal degradation in breast cancer cells. J Cell Biol 2018; 217:2777-2798. [PMID: 29980625 PMCID: PMC6080927 DOI: 10.1083/jcb.201703037] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 12/20/2017] [Accepted: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
Estrogen receptor α (ERα) is expressed in tissues as diverse as brains and mammary glands. In breast cancer, ERα is a key regulator of tumor progression. Therefore, understanding what activates ERα is critical for cancer treatment in particular and cell biology in general. Using biochemical approaches and superresolution microscopy, we show that estrogen drives membrane ERα into endosomes in breast cancer cells and that its fate is determined by the presence of fibronectin (FN) in the extracellular matrix; it is trafficked to lysosomes in the absence of FN and avoids the lysosomal compartment in its presence. In this context, FN prolongs ERα half-life and strengthens its transcriptional activity. We show that ERα is associated with β1-integrin at the membrane, and this integrin follows the same endocytosis and subcellular trafficking pathway triggered by estrogen. Moreover, ERα+ vesicles are present within human breast tissues, and colocalization with β1-integrin is detected primarily in tumors. Our work unravels a key, clinically relevant mechanism of microenvironmental regulation of ERα signaling.
Collapse
Affiliation(s)
- Rocío G Sampayo
- Universidad de Buenos Aires, Instituto de Oncología "Ángel H. Roffo", Área Investigación, Buenos Aires, Argentina .,Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Ciudad Universitaria, Buenos Aires, Argentina.,Universidad Nacional de San Martín, Instituto de Nanosistemas, Campus Miguelete, San Martín, Argentina
| | - Andrés M Toscani
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, IQUIBICEN UBA-CONICET y Universidad Nacional de Luján, Departamento de Ciencias Básicas, Buenos Aires, Argentina
| | - Matthew G Rubashkin
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Kate Thi
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Luciano A Masullo
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ianina L Violi
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Jonathon N Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Alfredo Cáceres
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, Córdoba, Argentina
| | - William C Hines
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Federico Coluccio Leskow
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, IQUIBICEN UBA-CONICET y Universidad Nacional de Luján, Departamento de Ciencias Básicas, Buenos Aires, Argentina
| | - Fernando D Stefani
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dante R Chialvo
- Center for Complex Systems and Brain Sciences, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Tecnológicas, San Martín, Argentina
| | - Mina J Bissell
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Marina Simian
- Universidad de Buenos Aires, Instituto de Oncología "Ángel H. Roffo", Área Investigación, Buenos Aires, Argentina .,Universidad Nacional de San Martín, Instituto de Nanosistemas, Campus Miguelete, San Martín, Argentina
| |
Collapse
|
5
|
Krzyscik M, Zakrzewska M, Sørensen V, Sokolowska-Wedzina A, Lobocki M, Swiderska KW, Krowarsch D, Wiedlocha A, Otlewski J. Cytotoxic Conjugates of Fibroblast Growth Factor 2 (FGF2) with Monomethyl Auristatin E for Effective Killing of Cells Expressing FGF Receptors. ACS OMEGA 2017; 2:3792-3805. [PMID: 30023704 PMCID: PMC6044718 DOI: 10.1021/acsomega.7b00116] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/28/2017] [Indexed: 06/08/2023]
Abstract
Antibody-drug conjugates (ADCs) are a new class of anticancer therapeutics that combine the selectivity of targeted treatment, ensured by monoclonal antibodies, with the potency of the cytotoxic agent. Here, we applied an analogous approach, but instead of an antibody, we used fibroblast growth factor 2 (FGF2). FGF2 is a natural ligand of fibroblast growth factor receptor 1 (FGFR1), a cell-surface receptor reported to be overexpressed in several types of tumors. We developed and characterized FGF2 conjugates containing a defined number of molecules of highly cytotoxic drug monomethyl auristatin E (MMAE). These conjugates effectively targeted FGFR1-expressing cells, were internalized upon FGFR1-mediated endocytosis, and, in consequence, revealed high cytotoxicity, which was clearly related to the FGFR1 expression level. Among the conjugates tested, the most potent was that bearing three MMAE molecules, showing that the cytotoxicity of protein-drug conjugates in vitro is directly dependent on drug loading.
Collapse
Affiliation(s)
- Mateusz
Adam Krzyscik
- Department
of Protein Engineering and Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Department
of Protein Engineering and Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Vigdis Sørensen
- Department of Core Facilities and Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Aleksandra Sokolowska-Wedzina
- Department
of Protein Engineering and Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Michal Lobocki
- Department
of Protein Engineering and Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Karolina Weronika Swiderska
- Department
of Protein Engineering and Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Daniel Krowarsch
- Department
of Protein Engineering and Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Antoni Wiedlocha
- Department of Core Facilities and Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Jacek Otlewski
- Department
of Protein Engineering and Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| |
Collapse
|
6
|
Bober J, Olsnes S, Kostas M, Bogacz M, Zakrzewska M, Otlewski J. Identification of new FGF1 binding partners-Implications for its intracellular function. IUBMB Life 2016; 68:242-51. [PMID: 26840910 PMCID: PMC4832500 DOI: 10.1002/iub.1480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/07/2016] [Indexed: 11/30/2022]
Abstract
Besides its classical mode of action through activation of specific receptors at the cell surface, fibroblast growth factor 1 (FGF1) can also cross the cellular membrane and translocate into the cytosol and further to the nucleus. The mechanism of this translocation is described partially, but the role of FGF1 inside the cell remains unknown. The aim of our work was to identify novel binding partners of FGF1 to predict its intracellular functions. We combined three methods of identification of such partners based on different principles: yeast two‐hybrid screen and mass spectrometry (MS) analysis of complexes obtained by Tandem Affinity Purification (TAP) or by co‐precipitation from cell lysate using recombinant FGF1. Altogether, we identified twenty novel intracellular proteins interacting with FGF1. For selected proteins, their direct interaction with FGF1 was confirmed by pull‐down assays and SPR measurements. Interestingly, half of the proteins found are involved in processes related to cell viability, such as apoptosis, cell proliferation, and cell cycle regulation. Thus, our study indicates that the role of intracellular FGF1 is to protect the cell against stress conditions by providing an additional signal for cell survival, independently of receptor‐activated signaling cascades. © 2016 IUBMB Life, 68(3):242–251, 2016
Collapse
Affiliation(s)
- Joanna Bober
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Wroclaw, Poland
| | - Sjur Olsnes
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Michal Kostas
- Faculty of Biotechnology, Department of Protein Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Marek Bogacz
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Wroclaw, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
7
|
Mueller NH, Fogueri U, Pedler MG, Montana K, Petrash JM, Ammar DA. Impact of Subunit Composition on the Uptake of α-Crystallin by Lens and Retina. PLoS One 2015; 10:e0137659. [PMID: 26355842 PMCID: PMC4565700 DOI: 10.1371/journal.pone.0137659] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/19/2015] [Indexed: 11/30/2022] Open
Abstract
Misfolded protein aggregation, including cataract, cause a significant amount of blindness worldwide. α-Crystallin is reported to bind misfolded proteins and prevent their aggregation. We hypothesize that supplementing retina and lens with α-crystallin may help to delay disease onset. The purpose of this study was to determine if αB-crystallin subunits containing a cell penetration peptide (gC-tagged αB-crystallin) facilitate the uptake of wild type αA-crystallin (WT-αA) in lens and retina. Recombinant human αB-crystallin was modified by the addition of a novel cell penetration peptide derived from the gC gene product of herpes simplex virus (gC-αB). Recombinant gC-αB and wild-type αA-crystallin (WT-αA) were purified from E. coli over-expression cultures. After Alexa-labeling of WT-αA, these proteins were mixed at ratios of 1:2, 1:5 and 1:10, respectively, and incubated at 37°C for 4 hours to allow for subunit exchange. Mixed oligomers were subsequently incubated with tissue culture cells or mouse organ cultures. Similarly, crystallin mixtures were injected into the vitreous of rat eyes. At various times after exposure, tissues were harvested and analyzed for protein uptake by confocal microscopy or flow cytometry. Chaperone-like activity assays were performed on α-crystallins ratios showing optimal uptake using chemically-induced or heat induced substrate aggregation assays. As determined by flow cytometry, a ratio of 1:5 for gC-αB to WT-αA was found to be optimal for uptake into retinal pigmented epithelial cells (ARPE-19). Chaperone-like activity assays demonstrated that hetero-oligomeric complex of gC-αB to WT-αA (in 1:5 ratio) retained protein aggregation protection. We observed a significant increase in protein uptake when optimized (gC-αB to WT-αA (1:5 ratio)) hetero-oligomers were used in mouse lens and retinal organ cultures. Increased levels of α-crystallin were found in lens and retina following intravitreal injection of homo- and hetero-oligomers in rats.
Collapse
Affiliation(s)
- Niklaus H. Mueller
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| | - Uma Fogueri
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Aurora, Colorado, United States of America
| | - Michelle G. Pedler
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Kameron Montana
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - J. Mark Petrash
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Aurora, Colorado, United States of America
| | - David A. Ammar
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
8
|
Mueller NH, Ammar DA, Petrash JM. Cell penetration peptides for enhanced entry of αB-crystallin into lens cells. Invest Ophthalmol Vis Sci 2013; 54:2-8. [PMID: 23150610 DOI: 10.1167/iovs.12-10947] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The prevalence of cataract increases with age. Conversely, the abundance of native α-crystallin diminishes with age and cataract development. We hypothesize replenishing lens α-crystallin may delay or prevent cataract. Herein we investigated the ability of cell penetration peptides (CPP) to enhance entry of α-crystallins into lens-derived cells. METHODS Recombinant αB-crystallins were modified by the addition of CPPs. Candidate CPP were designed with reference to the HSV-1 glycoprotein C gene (gC) or the HIV-1 TAT peptide. αB-crystallins produced by fusing gC or TAT were over-expressed in E. coli. Purified proteins were subjected to size exclusion chromatography (SEC) to characterize oligomeric complexes (OC). Chaperone-like activity (CLA) was evaluated by measuring the ability of α-crystallins to suppress chemically-induced protein aggregation. To evaluate protein uptake, labeled α-crystallins were incubated with HLE B3 cells and monitored by fluorescence microscopy for 48 hours. RESULTS We examined the effects of the addition of CPP on the structure, CLA, and cell transduction properties of αB-crystallins. C-terminal CPP fused crystallins had poor solubility. In contrast, N-terminal tagged αB-crystallins were soluble. These modified αB-crystallins formed OC that were larger than wild-type based on SEC. Wild-type and gC tagged αB-crystallin displayed robust CLA. Subunit exchange was observed when gC-fused αB-crystallin was mixed with αA. In contrast to wild-type, modified α-crystallins accumulated in HLE B3 cells. CONCLUSIONS Addition of CPP improves the uptake of αB-crystallins into HLE B3 cells. No undesirable changes to the chaperone-like abilities of α-crystallins were observed in αB-crystallin modified by the addition of the gC-derived CPP.
Collapse
Affiliation(s)
- Niklaus H Mueller
- Department of Ophthalmology, University of Colorado, School of Medicine, Aurora, CO, USA
| | | | | |
Collapse
|
9
|
Effect of the electrostatic potential on the internalization mechanism of cell penetrating peptides derived from TIRAP. BIOTECHNOL BIOPROC E 2012. [DOI: 10.1007/s12257-011-0482-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
10
|
Steroid-like signalling by interferons: making sense of specific gene activation by cytokines. Biochem J 2012; 443:329-38. [PMID: 22452815 DOI: 10.1042/bj20112187] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Many cytokines, hormones and growth factors use the JAK (Janus kinase)/STAT (signal transducer and activator of transcription) pathway for cell signalling and specific gene activation. In the classical model, ligand is said to interact solely with the receptor extracellular domain, which triggers JAK activation of STATs at the receptor cytoplasmic domain. Activated STATs are then said to carry out nuclear events of specific gene activation. Given the limited number of STATs (seven) and the activation of the same STATs by cytokines with different functions, the mechanism of the specificity of their signalling is not obvious. Focusing on IFNγ (interferon γ), we have shown that ligand, receptor and activated JAKs are involved in nuclear events that are associated with specific gene activation, where the receptor subunit IFNGR1 (IFNγ receptor 1) functions as a transcription/co-transcription factor and the JAKs are involved in key epigenetic events. RTKs (receptor tyrosine kinases) such as EGFR [EGF (epidermal growth factor) receptor] and FGFR [FGF (fibroblast growth factor) receptor] also undergo nuclear translocation in association with their respective ligands. EGFR and FGFR, like IFNGR1, have been shown to function as transcription/co-transcription factors. The RTKs also regulate other kinases that have epigenetic effects. Our IFNγ model, as well as the RTKs EGFR and FGFR, have similarities to that of steroid receptor signalling. These systems consist of ligand-receptor-co-activator complexes at the genes that they activate. The co-activators consist of transcription factors and kinases, of which the latter play an important role in the associated epigenetics. It is our view that signalling by cytokines such as IFNγ is but a variation of specific gene activation by steroid hormones.
Collapse
|
11
|
Suzuki A, Harada H, Nakamura H. Nuclear translocation of FGF8 and its implication to induce Sprouty2. Dev Growth Differ 2012; 54:463-73. [PMID: 22404534 DOI: 10.1111/j.1440-169x.2012.01332.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fibroblast growth factor 8 (FGF8) functions as a local organizing signal for the tectum and cerebellum. FGF8 activates Ras-ERK signaling pathway to induce cerebellar development. We paid attention to the difference in the expression pattern of the molecules that are induced by FGF8 in the mid-hind brain region during normal development and after FGF8 misexpression; some are expressed in the area corresponding to the ERK activation domain but the others are expressed corresponding to the Fgf8 expression domain. Since some of the FGF family members are localized in the nucleus, we wondered if FGF8 could localize in the nuclei and function in the nucleus. We first show that in cultured NIH3T3 cells transfected FGF8b could localize in the nucleus. Transfected FGF8b could also localize in the nucleus of the cells in the chick neural tube. In mouse embryonic neural tube, we detected endogenous FGF8 in the nuclei. Implantation of an FGF8b-soaked bead showed that exogenous FGF8b could be translocated to the nuclei in the isthmus. Furthermore, signal-peptide-deletion mutant of FGF8b mainly localized in the nuclei, and induced Sprouty2 without activating ERK in the mesencephalon. Signal-peptide-deletion mutant of FGF8b could not induce Pax2 expression. Taken together, we concluded that FGF8b could be translocated to the nuclei, and that the nuclear FGF8 could function as transcriptional regulator to induce Sprouty2 in the isthmus.
Collapse
Affiliation(s)
- Ayumu Suzuki
- Department of Molecular Neurobiology, Graduate School of Life Sciences and Institute of Development, Aging and Cancer, Tohoku University, Seiryo-machi 4-1, Aoba-ku, 980-8575 Sendai, Japan
| | | | | |
Collapse
|
12
|
Zhen Y, Sørensen V, Skjerpen CS, Haugsten EM, Jin Y, Wälchli S, Olsnes S, Wiedlocha A. Nuclear Import of Exogenous FGF1 Requires the ER-Protein LRRC59 and the Importins Kpnα1 and Kpnβ1. Traffic 2012; 13:650-64. [DOI: 10.1111/j.1600-0854.2012.01341.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 02/07/2012] [Accepted: 02/09/2012] [Indexed: 01/19/2023]
Affiliation(s)
| | | | | | | | | | - Sebastien Wälchli
- Department of Immunology; Institute for Cancer Research; The Norwegian Radium Hospital; Montebello; Oslo; 0310; Norway
| | | | | |
Collapse
|
13
|
Zwaenepoel O, Tzenaki N, Vergetaki A, Makrigiannakis A, Vanhaesebroeck B, Papakonstanti EA. Functional CSF-1 receptors are located at the nuclear envelope and activated via the p110δ isoform of PI 3-kinase. FASEB J 2011; 26:691-706. [PMID: 22084313 DOI: 10.1096/fj.11-189753] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Colony stimulating factor-1 (CSF-1) and its receptor (CSF-1R) are key regulators of macrophage biology, and their elevated expression in cancer cells has been linked to poor prognosis. CSF-1Rs are thought to function at the plasma membrane. We show here that functional CSF-1Rs are present at the nuclear envelope of various cell types, including primary macrophages, human cancer cell lines, and primary human carcinomas. In response to CSF-1, added to intact cells or isolated nuclei, nucleus-associated CSF-1R became phosphorylated and triggered the phosphorylation of Akt and p27 inside the nucleus. Extracellularly added CSF-1 was also found to colocalize with nucleus-associated CSF-1Rs. All these activities were found to depend selectively on the activity of the p110δ isoform of phosphoinositide 3-kinase (PI3K). This finding was related to the p110δ-dependent translocation of exogenous CSF-1 to the nucleus-associated CSF-1Rs, correlating with a prominent role of p110δ in activation of the Rab5 GTPase, a key regulator of the endocytic trafficking. siRNA-silencing of Rab5a phenocopied p110δ inactivation and nuclear CSF-1 signaling. Our work demonstrates for the first time the presence of functional nucleus-associated CSF-1Rs, which are activated by extracellular CSF-1 by a mechanism that involves p110δ and Rab5 activity. These findings may have important implications in cancer development.
Collapse
Affiliation(s)
- Olivier Zwaenepoel
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | | | | | | | | | | |
Collapse
|
14
|
Wang J, Liu XT, Huang H, Xiao G, Zhou ZY, Chen Y, Yu ZH, He SL, Chen AA, Wang DD, He Y, Zhang ZC, Hong A. Antitumor activity of a recombinant soluble ectodomain of mutant human fibroblast growth factor receptor-2 IIIc. Mol Cancer Ther 2011; 10:1656-66. [PMID: 21750221 DOI: 10.1158/1535-7163.mct-11-0163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The fibroblast growth factor (FGF) signaling pathway is a recognized target of cancer therapy. We have developed a strong inhibitor (S252W mutant soluble ectodomain of FGF recptor-2 IIIc, msFGFR2) that binds FGFs and blocks the activation of FGFRs. Thermodynamic binding studies indicated that msFGFR2 bound FGF-2 16.9 times as strongly as wild-type soluble FGFR2IIIc ectodomain (wsFGFR2). It successfully suppressed the growth, angiogenesis, and metastasis of two tumor cell lines in vitro and in vivo, and it potently inhibited cancer cell proliferation but not normal cell proliferation. Therefore, msFGFR2 is a useful probe for FGF-dependent signaling pathways and a potential broad-spectrum antitumor agent.
Collapse
Affiliation(s)
- Ju Wang
- Guangdong Provincial Key Laboratory of Bio-engineering Medicine, National Engineering Research Centre of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Zakrzewska M, Sørensen V, Jin Y, Wiedlocha A, Olsnes S. Translocation of exogenous FGF1 into cytosol and nucleus is a periodic event independent of receptor kinase activity. Exp Cell Res 2011; 317:1005-15. [DOI: 10.1016/j.yexcr.2011.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 12/10/2010] [Accepted: 01/03/2011] [Indexed: 10/18/2022]
|
16
|
Shi HL, Yang T, Deffar K, Dong CG, Liu JY, Fu CL, Zheng DX, Qin B, Wang JJ, Wang XZ, Zhu XJ. A novel single-chain variable fragment antibody against FGF-1 inhibits the growth of breast carcinoma cells by blocking the intracrine pathway of FGF-1. IUBMB Life 2011; 63:129-37. [PMID: 21360642 DOI: 10.1002/iub.423] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 12/27/2010] [Indexed: 11/05/2022]
Abstract
The fibroblast growth factors (FGFs) are important for embryo development, wound healing, hematopoiesis, and angiogenesis. FGF-1, a member of FGF family, is involved in both receptor-dependent pathways and an intracrine pathway. Studies have recently shown that FGF-1 is overexpressed in the early stages of several kinds of cancer. Thus, FGF-1 is a candidate for cancer immunotargeting. To study the potential use of therapeutic antibodies against FGF-1, a monoclonal hybridoma 1C9 secreting monoclonal antibody specific for FGF-1 was developed. Then, a single-chain variable fragment (scFv) antibody was genetically engineered from hybridama 1C9. The binding of the scFv1C9 to the antigen FGF-1 was demonstrated by ELISA and immunoprecipitation assays. Functional analysis showed that the overexpressed scFv1C9 in MCF-7 cells targeted endogenous FGF-1 and prevented the translocation of FGF-1 into the nucleus, resulting in the blockade of the intracrine pathway of FGF-1, which caused the G1 arrest by p21 up-regulation. These results suggest that the generated scFv1C9 is an effective inhibitor of the intracrine pathway of FGF-1 and has a potential application as anti-tumoral agent in breast cancer.
Collapse
Affiliation(s)
- Heng-Liang Shi
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zhou J, Liu W, Pong RC, Hao G, Sun X, Hsieh JT. Analysis of oligo-arginine cell-permeable peptides uptake by prostate cells. Amino Acids 2010; 42:1253-60. [PMID: 21120551 DOI: 10.1007/s00726-010-0817-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 11/18/2010] [Indexed: 12/24/2022]
Abstract
Recently, we have shown that oligo-arginine peptide (i.e., R11), a unique cell-permeable peptide (CPP), can be used as an imaging probe for prostate cancer detection. In this study, the mechanism(s) of oligo-arginine peptide in prostate cells was further analyzed. The length of the oligo-arginine peptide appears to be critical for the efficiency of uptake by prostate cells: poly (11)-arginine (R11) > poly (9)-arginine (R9) > poly (13)-arginine peptide (R13). The uptake of R11 peptide by prostate cells is mediated by macropinocytosis as evidenced by the fact that uptake can be blocked by a macropinocytosis inhibitor. However, the use of an inhibitor for carbohydrate chain elongation of glycosaminoglycan or inhibitors for carbohydrate synthesis of glycoprotein via either O-link or N-link showed minimal effects on R11 uptake. Nevertheless, pentosan sulfate (PentS) or dextran sulfate (DS) exhibited the highest inhibitory effect on R11 uptake in several prostate cells treated with various soluble glycosaminoglycans (GAGs) or anionic polymers. It is known that laminin receptor has been characterized as a PentS binding partner. Knocking down 37LRP (laminin receptor precursor) expression in prostate cells showed a reduction in their ability to uptake R11 peptides. In conclusion, laminin receptor is one of the initial binding site(s) responsible for R11 peptide uptake in prostate cells.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Urology, J8-134, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
18
|
Kubikova I, Konecna H, Sedo O, Zdrahal Z, Rehulka P, Hribkova H, Rehulkova H, Hampl A, Chmelik J, Dvorak P. Proteomic profiling of human embryonic stem cell-derived microvesicles reveals a risk of transfer of proteins of bovine and mouse origin. Cytotherapy 2009; 11:330-40, 1 p following 340. [PMID: 19401887 DOI: 10.1080/14653240802595531] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND AIMS Microvesicles (MV) shed from the plasma membrane of eukaryotic cells, including human embryonic stem cells (hESC), contain proteins, lipids and RNA and serve as mediators of cell-to-cell communication. However, they may also contain immunogenic membrane domains and infectious particles acquired from xenogenic components of the culture milieu. Therefore, MV represent a potential risk for clinical application of cell therapy. METHODS We tested the ability of hESC and their most commonly used feeder cells, mouse embryonic fibroblasts (MEF), to produce MV. We found that hESC are potent producers of MV, whereas mitotically inactivated MEF do not produce any detectable MV. We therefore employed a combined proteomic approach to identify the molecules that constitute the major components of MV from hESC maintained in a standard culture setting with xenogenic feeder cells. RESULTS In purified MV fractions, we identified a total of 22 proteins, including five unique protein species that are known to be highly expressed in invasive cancers and participate in cellular activation, metastasis and inhibition of apoptosis. Moreover, we found that hESC-derived MV contained the immunogenic agents apolipoprotein and transferrin, a source of Neu5Gc, as well as mouse retroviral Gag protein. CONCLUSIONS These findings indicate that MV represent a mechanism by which hESC communicate; however, they also serve as potential carriers of immunogenic and pathogenic compounds acquired from environment. Our results highlight a potential danger regarding the use of hESC that have previously been exposed to animal proteins and cells.
Collapse
Affiliation(s)
- I Kubikova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zong F, Fthenou E, Wolmer N, Hollósi P, Kovalszky I, Szilák L, Mogler C, Nilsonne G, Tzanakakis G, Dobra K. Syndecan-1 and FGF-2, but not FGF receptor-1, share a common transport route and co-localize with heparanase in the nuclei of mesenchymal tumor cells. PLoS One 2009; 4:e7346. [PMID: 19802384 PMCID: PMC2750749 DOI: 10.1371/journal.pone.0007346] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Accepted: 09/07/2009] [Indexed: 11/18/2022] Open
Abstract
Syndecan-1 forms complexes with growth factors and their cognate receptors in the cell membrane. We have previously reported a tubulin-mediated translocation of syndecan-1 to the nucleus. The transport route and functional significance of nuclear syndecan-1 is still incompletely understood. Here we investigate the sub-cellular distribution of syndecan-1, FGF-2, FGFR-1 and heparanase in malignant mesenchymal tumor cells, and explore the possibility of their coordinated translocation to the nucleus. To elucidate a structural requirement for this nuclear transport, we have transfected cells with a syndecan-1/EGFP construct or with a short truncated version containing only the tubulin binding RMKKK sequence. The sub-cellular distribution of the EGFP fusion proteins was monitored by fluorescence microscopy. Our data indicate that syndecan-1, FGF-2 and heparanase co-localize in the nucleus, whereas FGFR-1 is enriched mainly in the perinuclear area. Overexpression of syndecan-1 results in increased nuclear accumulation of FGF-2, demonstrating the functional importance of syndecan-1 for this nuclear transport. Interestingly, exogenously added FGF-2 does not follow the route taken by endogenous FGF-2. Furthermore, we prove that the RMKKK sequence of syndecan-1 is necessary and sufficient for nuclear translocation, acting as a nuclear localization signal, and the Arginine residue is vital for this localization. We conclude that syndecan-1 and FGF-2, but not FGFR-1 share a common transport route and co-localize with heparanase in the nucleus, and this transport is mediated by the RMKKK motif in syndecan-1. Our study opens a new perspective in the proteoglycan field and provides more evidence of nuclear interactions of syndecan-1.
Collapse
Affiliation(s)
- Fang Zong
- Department of Laboratory Medicine, Division of Pathology, Huddinge University Hospital, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Eleni Fthenou
- Department of Histology, Division of Morphology, School of Medicine, University of Crete, Heraklion, Greece
| | - Nina Wolmer
- Department of Laboratory Medicine, Division of Pathology, Huddinge University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Péter Hollósi
- 1st Institute of Pathology and Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - Ilona Kovalszky
- 1st Institute of Pathology and Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - László Szilák
- 1st Institute of Pathology and Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - Carolin Mogler
- Department of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Gustav Nilsonne
- Department of Laboratory Medicine, Division of Pathology, Huddinge University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Georgios Tzanakakis
- Department of Histology, Division of Morphology, School of Medicine, University of Crete, Heraklion, Greece
| | - Katalin Dobra
- Department of Laboratory Medicine, Division of Pathology, Huddinge University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Fibroblast growth factor-2 regulates the stability of nuclear bodies. Proc Natl Acad Sci U S A 2009; 106:12747-52. [PMID: 19617559 DOI: 10.1073/pnas.0900122106] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nuclear bodies are distinct subnuclear structures. The survival of motoneuron (SMN) gene is mutated or deleted in patients with the neurodegenerative disease spinal muscular atrophy (SMA). The gene product SMN is a marker protein for one class of nuclear bodies denoted as nuclear gems. SMN has also been found in Cajal bodies, which co-localize with gems in many cell types. Interestingly, SMA patients display a reduced number of gems. Little is known about the regulation of nuclear body formation and stabilization. We have previously shown that a nuclear isoform of the fibroblast growth factor-2 (FGF-2(23)) binds directly to SMN. In this study, we analyzed the consequences of FGF-2(23) binding to SMN with regard to nuclear body formation. On a molecular level, we showed that FGF-2(23) competed with Gemin2 (a component of the SMN complex that is necessary for gem stabilization) for binding to SMN. Down-regulation of Gemin2 by siRNA caused destabilization of SMN-positive nuclear bodies. This process is reflected in both cellular and in vivo systems by a negative regulatory function of FGF-2 in nuclear body formation: in HEK293 cells, FGF-2(23) decreased the number of SMN-positive nuclear bodies. The same effect could be observed in motoneurons of FGF-2 transgenic mice. This study demonstrates the functional role of a growth factor in the regulation of structural entities of the nucleus.
Collapse
|
21
|
Zakrzewska M, Wiedlocha A, Szlachcic A, Krowarsch D, Otlewski J, Olsnes S. Increased protein stability of FGF1 can compensate for its reduced affinity for heparin. J Biol Chem 2009; 284:25388-403. [PMID: 19574212 PMCID: PMC2757240 DOI: 10.1074/jbc.m109.001289] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human FGF1 (fibroblast growth factor 1) is a powerful signaling molecule with a short half-life in vivo and a denaturation temperature close to physiological. Binding to heparin increases the stability of FGF1 and is believed to be important in the formation of FGF1.fibroblast growth factor receptor (FGFR) active complex. In order to reveal the function of heparin in FGF1.FGFR complex formation and signaling, we constructed several FGF1 variants with reduced affinity for heparin and with diverse stability. We determined their biophysical properties and biological activities as well as their ability to translocate across cellular membranes. Our study showed that increased thermodynamic stability of FGF1 nicely compensates for decreased binding of heparin in FGFR activation, induction of DNA synthesis, and cell proliferation. By stepwise introduction of stabilizing mutations into the K118E (K132E) FGF1 variant that shows reduced affinity for heparin and is inactive in stimulation of DNA synthesis, we were able to restore the full mitogenic activity of this mutant. Our results indicate that the main role of heparin in FGF-induced signaling is to protect this naturally unstable protein against heat and/or proteolytic degradation and that heparin is not essential for a direct FGF1-FGFR interaction and receptor activation.
Collapse
Affiliation(s)
- Malgorzata Zakrzewska
- Centre for Cancer Biomedicine, University of Oslo, and Department of Biochemistry, Institute for Cancer Research, Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| | | | | | | | | | | |
Collapse
|
22
|
Zakrzewska M, Marcinkowska E, Wiedlocha A. FGF-1: From Biology Through Engineering to Potential Medical Applications. Crit Rev Clin Lab Sci 2008; 45:91-135. [DOI: 10.1080/10408360701713120] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
23
|
Stepanova V, Lebedeva T, Kuo A, Yarovoi S, Tkachuk S, Zaitsev S, Bdeir K, Dumler I, Marks MS, Parfyonova Y, Tkachuk VA, Higazi AAR, Cines DB. Nuclear translocation of urokinase-type plasminogen activator. Blood 2008; 112:100-10. [PMID: 18337556 PMCID: PMC2435680 DOI: 10.1182/blood-2007-07-104455] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 02/01/2008] [Indexed: 01/16/2023] Open
Abstract
Urokinase-type plasminogen activator (uPA) participates in diverse (patho)physiological processes through intracellular signaling events that affect cell adhesion, migration, and proliferation, although the mechanisms by which these occur are only partially understood. Here we report that upon cell binding and internalization, single-chain uPA (scuPA) translocates to the nucleus within minutes. Nuclear translocation does not involve proteolytic activation or degradation of scuPA. Neither the urokinase receptor (uPAR) nor the low-density lipoprotein-related receptor (LRP) is required for nuclear targeting. Rather, translocation involves the binding of scuPA to the nucleocytoplasmic shuttle protein nucleolin through a region containing the kringle domain. RNA interference and mutational analysis demonstrate that nucleolin is required for the nuclear transport of scuPA. Furthermore, nucleolin is required for the induction smooth muscle alpha-actin (alpha-SMA) by scuPA. These data reveal a novel pathway by which uPA is rapidly translocated to the nucleus where it might participate in regulating gene expression.
Collapse
Affiliation(s)
- Victoria Stepanova
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Phosphorylation of fibroblast growth factor (FGF) receptor 1 at Ser777 by p38 mitogen-activated protein kinase regulates translocation of exogenous FGF1 to the cytosol and nucleus. Mol Cell Biol 2008; 28:4129-41. [PMID: 18411303 DOI: 10.1128/mcb.02117-07] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Exogenous fibroblast growth factor 1 (FGF1) signals through activation of transmembrane FGF receptors (FGFRs) but may also regulate cellular processes after translocation to the cytosol and nucleus of target cells. Translocation of FGF1 occurs across the limiting membrane of intracellular vesicles and is a regulated process that depends on the C-terminal tail of the FGFR. Here, we report that translocation of FGF1 requires activity of the alpha isoform of p38 mitogen-activated protein kinase (MAPK). FGF1 translocation was inhibited after chemical inhibition of p38 MAPK or after small interfering RNA knockdown of p38alpha. Translocation was increased after stimulation of p38 MAPK with anisomycin, mannitol, or H2O2. The activity level of p38 MAPK was not found to affect endocytosis or intracellular sorting of FGF1/FGFR1. Instead, we found that p38 MAPK regulates FGF1 translocation by phosphorylation of FGFR1 at Ser777. The FGFR1 mutation S777A abolished FGF1 translocation, while phospho-mimetic mutations of Ser777 to Asp or Glu allowed translocation to take place and bypassed the requirement for active p38 MAPK. Ser777 in FGFR1 was directly phosphorylated by p38alpha in a cell-free system. These data demonstrate a crucial role for p38alpha MAPK in the regulated translocation of exogenous FGF1 into the cytosol/nucleus, and they reveal a specific role for p38alpha MAPK-mediated serine phosphorylation of FGFR1.
Collapse
|
25
|
Yu PJ, Ferrari G, Pirelli L, Galloway AC, Mignatti P, Pintucci G. Thrombin cleaves the high molecular weight forms of basic fibroblast growth factor (FGF-2): a novel mechanism for the control of FGF-2 and thrombin activity. Oncogene 2007; 27:2594-601. [PMID: 17968311 DOI: 10.1038/sj.onc.1210899] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The fgf-2 gene encodes low molecular weight (LMW, 18 kDa) and high molecular weight (HMW, 22-24 kDa) forms that originate from alternative translation of a single mRNA and exhibit diverse biological functions. HMW fibroblast growth factor-2 (FGF-2) inhibits cell migration and induces cell transformation or growth arrest in a cell type- and dose-dependent fashion. Conversely, LMW FGF-2 upregulates both cell proliferation and migration in most cell types. Although transcriptional and translational regulation of HMW and LMW FGF-2 has been extensively investigated, little is known about post-translational control of their relative expression. Here we report that thrombin, a key coagulation factor and inflammatory mediator, cleaves HMW FGF-2 into an LMW FGF-2-like form that stimulates endothelial cell migration and proliferation. The effect of thrombin on these cell functions requires HMW FGF-2 cleavage. This post-translational control mechanism adds a novel level of complexity to the regulation of FGF-2, and links the activities of thrombin and FGF-2 in patho-physiological processes in which both molecules are expressed.
Collapse
Affiliation(s)
- P-J Yu
- Department of Cardiothoracic Surgery, The Seymour Cohn Cardiovascular Research Laboratory, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | |
Collapse
|
26
|
Construction and characterization of a thrombin-resistant designer FGF-based collagen binding domain angiogen. Biomaterials 2007; 29:327-36. [PMID: 17950455 DOI: 10.1016/j.biomaterials.2007.09.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 09/23/2007] [Indexed: 11/21/2022]
Abstract
Humans demonstrate limited spontaneous endothelialization of prosthetic bypass grafts. However the local application of growth factors to prosthetic grafts or to injured blood vessels can provide an immediate effect on endothelialization. Novel chimeric proteins combining potent angiogens with extracellular matrix binding domains may localize to exposed matrices and provide sustained activity to promote endothelial regeneration after vascular interventions. We have ligated a thrombin-resistant mutant of fibroblast growth factor (FGF)-1 (R136K) with a collagen binding domain (CBD) in order to direct this growth factor to sites of exposed vascular collagen or selected bioengineered scaffolds. While FGF-1 and R136K are readily attracted to a variety of matrix proteins, R136K-CBD demonstrated selective and avid binding to collagen approximately 4x that of FGF-1 or R136K alone (P<0.05). The molecular stability of R136K-CBD was superior to FGF-1 and R136K. Its chemotactic activity was superior to R136K and FGF-1 (11+/-1% vs. 6+/-2% and 4+/-1%; P<0.01). Its angiogenic activity was similar to R136K and significantly greater than control by day 2 (P<0.01). After day 3, FGF-1-treated endothelial cell's (EC) sprouts had regressed back to levels insignificant compared to the control group (P=0.17), while both R136K and R136K-CBD continued to demonstrate greater sprout lengthening as compared to control (P<0.0002). The mitogenic activity of all growth factors was greater than control groups (20% PBS); in all comparisons (P<0.0001). This dual functioning angiogen provides proof of concept for the application of designer angiogens to matrix binding proteins to intelligently promote endothelial regeneration of selected matrices.
Collapse
|
27
|
Nilsen T, Rosendal KR, Sørensen V, Wesche J, Olsnes S, Wiedłocha A. A nuclear export sequence located on a beta-strand in fibroblast growth factor-1. J Biol Chem 2007; 282:26245-56. [PMID: 17616529 DOI: 10.1074/jbc.m611234200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Receptor-bound and endocytosed fibroblast growth factor-1 (FGF-1) is able to cross the vesicle membrane and translocate to cytosol and nucleus. This suggests an intracellular role of FGF-1, which also signals by activating transmembrane FGF receptors. Phosphorylation of internalized FGF-1 by nuclear protein kinase C delta induces rapid export from the nuclei by a leptomycin B-sensitive pathway. In the present work, we have searched for and identified a Leu-rich nuclear export sequence (NES) at the C terminus of FGF-1 required for its nuclear export and able to confer nuclear export activity to a reporter protein in an in vivo system. Mutants where hydrophobic amino acids within the NES were exchanged for alanine exhibited reduced or abolished nuclear export. As demonstrated in co-immunoprecipitation experiments, a complex containing FGF-1, exportin-1, and its co-factor Ran-GTP, was formed in vitro. Formation of this complex in vivo was demonstrated by a peroxisomal targeting assay. Formation of the FGF-1-exportin-1-Ran-GTP complex in vitro as well as nuclear export of FGF-1 in vivo was dependent on phosphorylation of FGF-1, and it was abolished by leptomycin B. The FGF-1 NES was found to be situated along a beta-strand, which has not been reported before, since NESs usually are alpha-helical.
Collapse
Affiliation(s)
- Trine Nilsen
- Centre for Cancer Biomedicine, Institute for Cancer Research, Rikshospitalet-Radiumhospitalet Medical Centre, Montebello, University of Oslo, 0310 Oslo, Norway
| | | | | | | | | | | |
Collapse
|
28
|
Duchardt F, Fotin-Mleczek M, Schwarz H, Fischer R, Brock R. A comprehensive model for the cellular uptake of cationic cell-penetrating peptides. Traffic 2007; 8:848-66. [PMID: 17587406 DOI: 10.1111/j.1600-0854.2007.00572.x] [Citation(s) in RCA: 628] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The plasma membrane represents an impermeable barrier for most macromolecules. Still some proteins and so-called cell-penetrating peptides enter cells efficiently. It has been shown that endocytosis contributes to the import of these molecules. However, conflicting results have been obtained concerning the nature of the endocytic process. In addition, there have been new findings for an endocytosis-independent cellular entry. In this study, we provide evidence that the Antennapedia-homeodomain-derived antennapedia (Antp) peptide, nona-arginine and the HIV-1 Tat-protein-derived Tat peptide simultaneously use three endocytic pathways: macropinocytosis, clathrin-mediated endocytosis and caveolae/lipid-raft-mediated endocytosis. Antennapedia differs from Tat and R9 by the extent by which the different import mechanisms contribute to uptake. Moreover, at higher concentrations, uptake occurs by a mechanism that originates from spatially restricted sites of the plasma membrane and leads to a rapid cytoplasmic distribution of the peptides. Endocytic vesicles could not be detected, suggesting an endocytosis-independent mode of uptake. Heparinase treatment of cells negatively affects this import, as does the protein kinase C inhibitor rottlerin, expression of dominant-negative dynamin and chlorpromazine. This mechanism of uptake was observed for a panel of different cell lines. For Antp, significantly higher peptide concentrations and inhibition of endocytosis were required to induce its uptake. The relevance of these findings for import of biologically active cargos is shown.
Collapse
Affiliation(s)
- Falk Duchardt
- Interfaculty Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | | | | | | | | |
Collapse
|
29
|
Herskovits AA, Auerbuch V, Portnoy DA. Bacterial ligands generated in a phagosome are targets of the cytosolic innate immune system. PLoS Pathog 2007; 3:e51. [PMID: 17397264 PMCID: PMC1839167 DOI: 10.1371/journal.ppat.0030051] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Accepted: 02/16/2007] [Indexed: 12/13/2022] Open
Abstract
Macrophages are permissive hosts to intracellular pathogens, but upon activation become microbiocidal effectors of innate and cell-mediated immunity. How the fate of internalized microorganisms is monitored by macrophages, and how that information is integrated to stimulate specific immune responses is not understood. Activation of macrophages with interferon (IFN)–γ leads to rapid killing and degradation of Listeria monocytogenes in a phagosome, thus preventing escape of bacteria to the cytosol. Here, we show that activated macrophages induce a specific gene expression program to L. monocytogenes degraded in the phago-lysosome. In addition to activation of Toll-like receptor (TLR) signaling pathways, degraded bacteria also activated a TLR-independent transcriptional response that was similar to the response induced by cytosolic L. monocytogenes. More specifically, degraded bacteria induced a TLR-independent IFN-β response that was previously shown to be specific to cytosolic bacteria and not to intact bacteria localized to the phagosome. This response required the generation of bacterial ligands in the phago-lysosome and was largely dependent on nucleotide-binding oligomerization domain 2 (NOD2), a cytosolic receptor known to respond to bacterial peptidoglycan fragments. The NOD2-dependent response to degraded bacteria required the phagosomal membrane potential and the activity of lysosomal proteases. The NOD2-dependent IFN-β production resulted from synergism with other cytosolic microbial sensors. This study supports the hypothesis that in activated macrophages, cytosolic innate immune receptors are activated by bacterial ligands generated in the phagosome and transported to the cytosol. Innate immune recognition of microorganisms has a direct impact on the type and the magnitude of the immune response elicited. While recognition of microorganisms relies on receptors that sense pathogen-associated molecular patterns, (PAMPs), it was reasonable to suspect that immune cells could discriminate between live and dead bacteria. Listeria monocytogenes is an intracellular pathogenic bacterium used extensively as a model system for studying basic aspects of innate and acquired immunity. L. monocytogenes is internalized by macrophages, escapes from a vacuole, multiplies within the cytosol, and spreads from cell to cell without lysing the cells. We used wild-type and bacterial mutants of L. monocytogenes to demonstrate that macrophages not only respond differently to bacteria that are growing in the cytosol and to non-growing bacteria that are trapped in a vacuole, but that they also can discriminate between intact or degraded bacteria in the vacuole. We showed that macrophages induce specific immune response when bacteria are killed and degraded. This response was directly correlated to the ability of macrophages to degrade bacteria and involved receptors that were located in the host cell cytosol. These observations led us to suggest that bacterial degradation products may serve as messengers that inform immune cells that bacteria were killed and degraded. This information might affect directly the immune response, for example, by down-regulating inflammatory responses that can be deleterious. We call these bacterial degradation products PAMP-PM (PAMP–post-mortem).
Collapse
Affiliation(s)
- Anat A Herskovits
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Victoria Auerbuch
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- School of Public Health, University of California, Berkeley, California, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
30
|
Planque N. Nuclear trafficking of secreted factors and cell-surface receptors: new pathways to regulate cell proliferation and differentiation, and involvement in cancers. Cell Commun Signal 2006; 4:7. [PMID: 17049074 PMCID: PMC1626074 DOI: 10.1186/1478-811x-4-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Accepted: 10/18/2006] [Indexed: 12/14/2022] Open
Abstract
Secreted factors and cell surface receptors can be internalized by endocytosis and translocated to the cytoplasm. Instead of being recycled or proteolysed, they sometimes translocate to the nucleus. Nuclear import generally involves a nuclear localization signal contained either in the secreted factor or its transmembrane receptor, that is recognized by the importins machinery. In the nucleus, these molecules regulate transcription of specific target genes by direct binding to transcription factors or general coregulators. In addition to the transcription regulation, nuclear secreted proteins and receptors seem to be involved in other important processes for cell life and cellular integrity such as DNA replication, DNA repair and RNA metabolism. Nuclear secreted proteins and transmembrane receptors now appear to induce new signaling pathways to regulate cell proliferation and differentiation. Their nuclear localization is often transient, appearing only during certain phases of the cell cycle. Nuclear secreted and transmembrane molecules regulate the proliferation and differentiation of a large panel of cell types during embryogenesis and adulthood and are also potentially involved in wound healing. Secreted factors such as CCN proteins, EGF, FGFs and their receptors are often detected in the nucleus of cancer cells. Nuclear localization of these molecules has been correlated with tumor progression and poor prognosis for patient survival. Nuclear growth factors and receptors may be responsible for resistance to radiotherapy.
Collapse
Affiliation(s)
- Nathalie Planque
- Laboratoire d'Oncologie Virale et Moléculaire, Université Paris7-Denis Diderot, UFR de Biochimie, 2 place Jussieu, 75005 Paris, France.
| |
Collapse
|
31
|
Sørensen V, Wiedlocha A, Haugsten EM, Khnykin D, Wesche J, Olsnes S. Different abilities of the four FGFRs to mediate FGF-1 translocation are linked to differences in the receptor C-terminal tail. J Cell Sci 2006; 119:4332-41. [PMID: 17003104 DOI: 10.1242/jcs.03209] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Members of the fibroblast growth factor family bind to one or more of the four closely related membrane-spanning FGF receptors. In addition to signaling through the receptors, exogenous FGF-1 and FGF-2 are endocytosed and translocated to the cytosol and nucleus where they stimulate RNA and DNA synthesis. Here we have studied the ability of the four FGF receptors to facilitate translocation of exogenous FGF-1 to the cytosol and nucleus. FGFR1 and FGFR4 were able to mediate translocation, whereas FGFR2 and FGFR3 completely lacked this ability. By analyzing mutant FGFRs we found that the tyrosine kinase domain could be deleted from FGFR1 without abolishing translocation, whereas the C-terminal tail of the FGFRs, constituted by approximately 50 amino acids downstream of the kinase domain, plays a crucial role in FGF-1 translocation. Three amino acids residues within the C-terminal tail were found to be of particular importance for translocation. For FGFR2, the two amino acid substitutions Q774M and P800H were sufficient to enable the receptor to support FGF-1 translocation. The results demonstrate a striking diversity in function of the four FGFRs determined by their C-terminal domain.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blotting, Western
- COS Cells
- Cattle
- Chlorocebus aethiops
- Electrophoresis, Polyacrylamide Gel
- Fibroblast Growth Factor 1/metabolism
- HeLa Cells
- Humans
- Kinetics
- Mice
- Molecular Sequence Data
- Mutation/genetics
- Phosphorylation
- Protein Transport/physiology
- Rats
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/physiology
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/physiology
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Receptor, Fibroblast Growth Factor, Type 3/physiology
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Receptor, Fibroblast Growth Factor, Type 4/physiology
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Receptors, Fibroblast Growth Factor/physiology
- Sequence Homology, Amino Acid
Collapse
Affiliation(s)
- Vigdis Sørensen
- The Department of Biochemistry, Institute for Cancer Research, The University of Oslo, The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
32
|
Dupree MA, Pollack SR, Levine EM, Laurencin CT. Fibroblast growth factor 2 induced proliferation in osteoblasts and bone marrow stromal cells: a whole cell model. Biophys J 2006; 91:3097-112. [PMID: 16861274 PMCID: PMC1578487 DOI: 10.1529/biophysj.106.087098] [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: 11/18/2022] Open
Abstract
Fibroblast growth factor 2 (FGF2) can enhance the proliferative capacity of bone and bone marrow stromal cells; however, the mechanisms behind this effect are not well described. We present a whole-cell kinetic model relating receptor-mediated binding, internalization, and processing of FGF2 to osteoblastic proliferative response. Focusing on one of the potential signaling complex stoichiometries, we utilized experimentally measured and modeled estimated rate constants to predict in vitro proliferation and distinguish between potential binding orders. We found that piecewise assemblage of a ternary signaling complex may occur in several ways depending on the local binding environment. Using experimental data of endocytosed FGF2 as a constraint, we have also shown evidence of potential multistep processes involved in heparan-sulfate proteoglycans-bound FGF2 release, internalization, and fragment formation in conjunction with the normal metabolism of the proteoglycan.
Collapse
Affiliation(s)
- Melissa A Dupree
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
33
|
Sørensen V, Nilsen T, Wiedłocha A. Functional diversity of FGF-2 isoforms by intracellular sorting. Bioessays 2006; 28:504-14. [PMID: 16615083 DOI: 10.1002/bies.20405] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Regulation of the subcellular localization of certain proteins is a mechanism for the regulation of their biological activities. FGF-2 can be produced as distinct isoforms by alternative initiation of translation on a single mRNA and the isoforms are differently sorted in cells. High molecular weight FGF-2 isoforms are not secreted from the cell, but are transported to the nucleus where they regulate cell growth or behavior in an intracrine fashion. 18 kDa FGF-2 can be secreted to the extracellular medium where it acts as a conventional growth factor by binding to and activation of cell-surface receptors. Furthermore, following receptor-mediated endocytosis, the exogenous FGF-2 can be transported to the nuclei of target cells, and this is of importance for the transmittance of a mitogenic signal. The growth factor is able to interact with several intracellular proteins. Here, the mode of action and biological role of intracellular FGF-2 are discussed.
Collapse
Affiliation(s)
- Vigdis Sørensen
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, University of Oslo, Norway
| | | | | |
Collapse
|
34
|
Wesche J, Małecki J, Wiedłocha A, Skjerpen CS, Claus P, Olsnes S. FGF-1 and FGF-2 Require the Cytosolic Chaperone Hsp90 for Translocation into the Cytosol and the Cell Nucleus. J Biol Chem 2006; 281:11405-12. [PMID: 16495214 DOI: 10.1074/jbc.m600477200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Similarly to many protein toxins, the growth factors fibroblast growth factor 1 (FGF-1) and FGF-2 translocate from endosomes into the cytosol. It was recently found that certain toxins are dependent on cytosolic Hsp90 for efficient translocation across the endosomal membrane. We therefore investigated the requirement for Hsp90 in FGF translocation. We found that low concentrations of the specific Hsp90 inhibitors, geldanamycin and radicicol, completely blocked the translocation of FGF-1 and FGF-2 to the cytosol and the nucleus. The drugs did not interfere with the initial binding of FGF-1 to the growth factor receptors at the cell-surface or with the subsequent internalization of the growth factors into endosomes. The activation of known signaling cascades downstream of the growth factor receptors was also not affected by the drugs. The data indicate that the drugs block translocation from endosomes to the cytosol implying that Hsp90 is required for translocation of FGF-1 and FGF-2 across the endosomal membrane.
Collapse
Affiliation(s)
- Jørgen Wesche
- Institute for Cancer Research at the Norwegian Radium Hospital, University of Oslo, Montebello, 0310 Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
35
|
Reigstad LJ, Martinez A, Varhaug JE, Lillehaug JR. Nuclear localisation of endogenous SUMO-1-modified PDGF-C in human thyroid tissue and cell lines. Exp Cell Res 2006; 312:782-95. [PMID: 16443219 DOI: 10.1016/j.yexcr.2005.11.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Revised: 11/21/2005] [Accepted: 11/30/2005] [Indexed: 01/06/2023]
Abstract
We investigated post-translational modification and subcellular localisation of endogenous platelet-derived growth factor-C (PDGF-C) in human thyroid papillary carcinomas (PTC), non-neoplastic thyroid tissues, and a selection of cultured cell lines. PDGF-C expressed nuclear localisation in 95% of all tested cell types in culture and in 10% of the thyrocytes from both PTC and non-neoplastic tissue. The cell lines expressed two forms of full-length PDGF-C, approximately 39 and approximately 55 kDa, in cell membrane and cytosol, while the approximately 55 kDa form dominated in the nucleus where it was partly chromatin-associated. The approximately 55 kDa form was post-translationally modified by SUMO-1. The putative PDGF-C SUMOylation site is the surface exposed (314)lysine part of a positively charged loop ((312)RPKTGVRGLHK(322)) with characteristics of a nuclear localisation signal. The tissue thyrocytes expressed a non-SUMOylated approximately 43 kDa and the 55 kDa PDGF-C. The SUMO-1 modified approximately 55 kDa PDGF-C expression was low in PTC where the approximately 43 kDa PDGF-C dominated. This is in contrast to non-neoplastic tissue and cultured cells where the SUMOylated approximately 55 kDa PDGF-C was strongly expressed. Our data provide novel evidence for nuclear localisation of PDGF-C, post-translational modification by SUMOylation and the expression of a novel form of PDGF-C in human papillary thyroid carcinomas.
Collapse
Affiliation(s)
- Laila J Reigstad
- Department of Molecular Biology, University of Bergen, Section of Surgery, Haukeland University Hospital, Bergen 5020, Norway
| | | | | | | |
Collapse
|
36
|
Abstract
First identified as peptides derived from the human immunodeficiency virus (HIV) transcriptional regulator Tat and the Drosophila transcription factor Antennapedia, transduction (or cell-penetrating) peptide sequences enable soluble proteins to cross biological membranes and interact with cytosolic and nuclear targets. Proteins containing such sequences have been found to function as transcription factors, to inhibit apoptosis, to play roles in axon guidance, or to transport viral mRNA between cells. The recent demonstration that dynorphins are able to act as transduction peptides suggests that these neuropeptides may have roles independent of opiate receptor activation.
Collapse
Affiliation(s)
- Alain Joliot
- Homeoprotein Cell Biology, CNRS UMR 8542, Ecole Normale Supérieure, Paris, France.
| |
Collapse
|
37
|
Fischer R, Fotin-Mleczek M, Hufnagel H, Brock R. Break on through to the Other Side-Biophysics and Cell Biology Shed Light on Cell-Penetrating Peptides. Chembiochem 2005; 6:2126-42. [PMID: 16254940 DOI: 10.1002/cbic.200500044] [Citation(s) in RCA: 212] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cell-penetrating peptides (CPPs) have become widely used vectors for the cellular import of molecules in basic and applied biomedical research. Despite the broad acceptance of these molecules as molecular carriers, the details of the mode of cellular internalization and membrane permeation remain elusive. Within the last two years endocytosis has been demonstrated to be a route of uptake shared by several CPPs. These findings had a significant impact on CPP research. State-of-the-art cell biology is now required to advance the understanding of the intracellular fate of the CPP and cargo molecules. Owing to their presumed ability to cross lipid bilayers, CPPs also represent highly interesting objects of biophysical research. Numerous studies have investigated structure-activity relationships of CPPs with respect to their ability to bind to a lipid bilayer or to cross this barrier. Endocytosis route only relocates the membrane permeation from the cell surface to endocytic compartments. Therefore, biophysical experiments are key to a mechanistic molecular understanding of the cellular uptake of CPPs. However, biophysical investigations have to consider the molecular environment encountered by a peptide inside and outside a cell. In this contribution we will review biophysical and cell-biology data obtained for several prominent CPPs. Furthermore, we will summarize recent findings on the cell-penetrating characteristics of antimicrobial peptides and the antimicrobial properties of CPPs. Peptides of both groups have overlapping characteristics. Therefore, both fields may greatly benefit from each other. The review will conclude with a perspective of how biophysics and cell biology may synergize even more efficiently in the future.
Collapse
Affiliation(s)
- Rainer Fischer
- Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen,, Germany
| | | | | | | |
Collapse
|
38
|
Abstract
Malignant tumours can spread to lymph nodes through lymphatic vessels. Recent studies show that tumours produce a range of growth factors that directly or indirectly stimulate lymphatic vessel growth (lymphangiogenesis) and lymphatic metastasis. These findings indicate that tumour lymphangiogenesis, similar to haemangiogenesis, is a complex process that is regulated by multiple growth factors. Understanding the underlying mechanisms by which tumours induce lymphangiogenesis might provide important information for the therapeutic intervention of metastatic spread.
Collapse
Affiliation(s)
- Yihai Cao
- Laboratory of Angiogenesis Research, Microbiology and Tumour Biology Center, Karolinska Institute, 171 77 Stockholm, Sweden.
| |
Collapse
|
39
|
Abstract
The nuclear localization of a number of growth factors, cytokine ligands and their receptors has been reported in various cell lines and tissues. These include members of the fibroblast growth factor (FGF), epidermal growth factor and growth hormone families. Accordingly, a number of nuclear functions have begun to emerge for these protein families. The demonstration of functional interactions of these proteins with the nuclear import machinery has further supported their functions as nuclear signal transducers. Here, we review the membrane- trafficking machinery and pathways demonstrated to regulate this cell surface to nucleus-trafficking event and highlight the many remaining unanswered questions. We focus on the FGF family, which is providing many of the clues as to the process of this unusual phenomenon.
Collapse
Affiliation(s)
- David M Bryant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | | |
Collapse
|
40
|
Gringel S, van Bergeijk J, Haastert K, Grothe C, Claus P. Nuclear fibroblast growth factor-2 interacts specifically with splicing factor SF3a66. Biol Chem 2005; 385:1203-8. [PMID: 15653435 DOI: 10.1515/bc.2004.156] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Fibroblast growth factor 2 (FGF-2) has a dual role as a classical extracellular signaling protein and as an intracellular factor. Isoforms of FGF-2, resulting from alternatively used start codons on one mRNA species, locate differentially to nuclear compartments. In this study we aimed to analyze functions of intracellular FGF-2 by identification of interacting proteins. We identified the 66-kDa subunit of splicing factor 3a (SF3a66) as a binding partner in a yeast two-hybrid screen and confirmed this interaction by pull-down assays. The splicing factor interacted with the 18-kDa (FGF-2(18)) and with the 23-kDa (FGF-2(23)) isoforms, indicating an interaction with a domain common to both isoforms. Moreover, FGF-2 interacted with the C-terminus of SF3a66, a sequence that has not previously been assigned a functional role. In a functional neurite outgrowth assay, SF3a66 enhanced neurite lengths similar to FGF-2(18). We have previously identified the spliceosomal assembly factor survival of motoneuron (SMN) protein as a protein interacting specifically with the FGF-2(23) isoform [Claus et al., J. Biol. Chem. 278 (2003), 479-485]. The identification of two FGF-2 interacting proteins from the same biochemical pathway suggests a novel intranuclear role of FGF-2.
Collapse
Affiliation(s)
- Susanne Gringel
- Department of Neuroanatomy, Hannover Medical School, D-30625 Hannover, Germany
| | | | | | | | | |
Collapse
|
41
|
Millette E, Rauch BH, Defawe O, Kenagy RD, Daum G, Clowes AW. Platelet-Derived Growth Factor-BB–Induced Human Smooth Muscle Cell Proliferation Depends on Basic FGF Release and FGFR-1 Activation. Circ Res 2005; 96:172-9. [PMID: 15625285 DOI: 10.1161/01.res.0000154595.87608.db] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have shown that the G protein–coupled receptor (GPCR) agonists, thrombin and Factor Xa, stimulate smooth muscle cell (SMC) proliferation through transactivation of the EGF receptor (EGFR) or the FGF receptor (FGFR), both of which are tyrosine kinase receptors. In the present study, we investigated whether platelet-derived growth factor (PDGF), a tyrosine kinase receptor agonist, might transactivate another tyrosine kinase receptor to induce SMC proliferation. Because heparin inhibits PDGF-mediated proliferation in human SMCs, we investigated whether the heparin-binding growth factor basic fibroblast growth factor (bFGF) and one of its receptors, FGFR-1, play a role in the response of human arterial SMCs to PDGF-BB. PDGF-BB induced the release of bFGF and sustained phosphorylation of FGFR-1 (30 minutes to 6 hours). A bFGF-neutralizing antibody inhibited PDGF-BB–mediated phosphorylation of FGFR-1, DNA synthesis, and cell proliferation. In the presence of bFGF antibody, PDGF-BB–induced early activation of ERK (0 to 60 minutes) was not affected, whereas late ERK activation (2 to 4 hours) was reduced. When FGFR-1 expression was suppressed using small interfering RNA (siRNA), ERK activation was reduced at late, but not early, time points after PDGF-BB stimulation. Addition of bFGF antibody to cells treated with siRNA to FGFR-1 had no further effect on ERK activation. Our results provide support for a novel mechanism by which PDGF-BB induces the release of bFGF and activation of FGFR-1 followed by the sustained activation of ERK and proliferation of human SMCs.
Collapse
MESH Headings
- Aorta, Abdominal
- Becaplermin
- Cell Division/drug effects
- Cell Movement/drug effects
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Chromones/pharmacology
- DNA Replication/drug effects
- Enzyme Activation/drug effects
- Fibroblast Growth Factor 2/metabolism
- Fibroblast Growth Factor 2/pharmacology
- Fibroblast Growth Factor 2/physiology
- Flavonoids/pharmacology
- Heparin/pharmacology
- Humans
- Indoles/pharmacology
- MAP Kinase Kinase 1/metabolism
- MAP Kinase Kinase 2/metabolism
- Maleimides/pharmacology
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Morpholines/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phosphorylation/drug effects
- Platelet-Derived Growth Factor/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Protein Processing, Post-Translational/drug effects
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- Proto-Oncogene Proteins c-sis
- RNA, Small Interfering/pharmacology
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/physiology
- Receptor, Fibroblast Growth Factor, Type 1
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/physiology
- Recombinant Proteins/pharmacology
- Tyrphostins/pharmacology
Collapse
Affiliation(s)
- Esther Millette
- University of Washington School of Medicine, Department of Surgery, Box 356410, 1959 NE Pacific St, Seattle, WA 98195-6410, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Wiedłocha A, Nilsen T, Wesche J, Sørensen V, Małecki J, Marcinkowska E, Olsnes S. Phosphorylation-regulated nucleocytoplasmic trafficking of internalized fibroblast growth factor-1. Mol Biol Cell 2004; 16:794-810. [PMID: 15574884 PMCID: PMC545912 DOI: 10.1091/mbc.e04-05-0389] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Fibroblast growth factor-1 (FGF-1), which stimulates cell growth, differentiation, and migration, is capable of crossing cellular membranes to reach the cytosol and the nucleus in cells containing specific FGF receptors. The cell entry process can be monitored by phosphorylation of the translocated FGF-1. We present evidence that phosphorylation of FGF-1 occurs in the nucleus by protein kinase C (PKC)delta. The phosphorylated FGF-1 is subsequently exported to the cytosol. A mutant growth factor where serine at the phosphorylation site is exchanged with glutamic acid, to mimic phosphorylated FGF-1, is constitutively transported to the cytosol, whereas a mutant containing alanine at this site remains in the nucleus. The export can be blocked by leptomycin B, indicating active and receptor-mediated nuclear export of FGF-1. Thapsigargin, but not leptomycin B, prevents the appearance of active PKCdelta in the nucleus, and FGF-1 is in this case phosphorylated in the cytosol. Leptomycin B increases the amount of phosphorylated FGF-1 in the cells by preventing dephosphorylation of the growth factor, which seems to occur more rapidly in the cytoplasm than in the nucleus. The nucleocytoplasmic trafficking of the phosphorylated growth factor is likely to play a role in the activity of internalized FGF-1.
Collapse
Affiliation(s)
- Antoni Wiedłocha
- Institute for Cancer Research, The Norwegian Radium Hospital, 0310 Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|