1
|
Bogale DE. The roles of FGFR3 and c-MYC in urothelial bladder cancer. Discov Oncol 2024; 15:295. [PMID: 39031286 PMCID: PMC11264706 DOI: 10.1007/s12672-024-01173-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/16/2024] [Indexed: 07/22/2024] Open
Abstract
Bladder cancer is one of the most frequently occurring cancers worldwide. At diagnosis, 75% of urothelial bladder cancer cases have non-muscle invasive bladder cancer while 25% have muscle invasive or metastatic disease. Aberrantly activated fibroblast growth factor receptor (FGFR)-3 has been implicated in the pathogenesis of bladder cancer. Activating mutations of FGFR3 are observed in around 70% of NMIBC cases and ~ 15% of MIBCs. Activated FGFR3 leads to ligand-independent receptor dimerization and activation of downstream signaling pathways that promote cell proliferation and survival. FGFR3 is an important therapeutic target in bladder cancer, and clinical studies have shown the benefit of FGFR inhibitors in a subset of bladder cancer patients. c-MYC is a well-known major driver of carcinogenesis and is one of the most commonly deregulated oncogenes identified in human cancers. Studies have shown that the antitumor effects of FGFR inhibition in FGFR3 dependent bladder cancer cells and other FGFR dependent cancers may be mediated through c-MYC, a key downstream effector of activated FGFR that is involved tumorigenesis. This review will summarize the current general understanding of FGFR signaling and MYC alterations in cancer, and the role of FGFR3 and MYC dysregulation in the pathogenesis of urothelial bladder cancer with the possible therapeutic implications.
Collapse
Affiliation(s)
- Dereje E Bogale
- School of Medicine, Department of Oncology, Addis Ababa University, Addis Ababa, Ethiopia.
| |
Collapse
|
2
|
Pande S, Vary C, Yang X, Liaw L, Gower L, Friesel R, Prudovsky I, Ryzhov S. Endothelial IL17RD promotes Western diet-induced aortic myeloid cell infiltration. Biochem Biophys Res Commun 2024; 701:149552. [PMID: 38335918 PMCID: PMC10936543 DOI: 10.1016/j.bbrc.2024.149552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024]
Abstract
The Interleukin-17 (IL17) family is a group of cytokines implicated in the etiology of several inflammatory diseases. Interleukin-17 receptor D (IL17RD), also known as Sef (similar expression to fibroblast growth factor) belonging to the family of IL17 receptors, has been shown to modulate IL17A-associated inflammatory phenotypes. The objective of this study was to test the hypothesis that IL17RD promotes endothelial cell activation and consequent leukocyte adhesion. We utilized primary human aortic endothelial cells and demonstrated that RNAi targeting of IL17RD suppressed transcript levels by 83 % compared to non-targeted controls. Further, RNAi knockdown of IL17RD decreased the adhesion of THP-1 monocytic cells onto a monolayer of aortic endothelial cells in response to IL17A. Additionally, we determined that IL17A did not significantly enhance the activation of canonical MAPK and NFκB pathways in endothelial cells, and further did not significantly affect the expression of VCAM-1 and ICAM-1 in aortic endothelial cells, which is contrary to previous findings. We also determined the functional relevance of our findings in vivo by comparing the expression of endothelial VCAM-1 and ICAM-1 and leukocyte infiltration in the aorta in Western diet-fed Il17rd null versus wild-type mice. Our results showed that although Il17rd null mice do not have significant alteration in aortic expression of VCAM-1 and ICAM-1 in endothelial cells, they exhibit decreased accumulation of proinflammatory monocytes and neutrophils, suggesting that endothelial IL17RD induced in vivo myeloid cell accumulation is not dependent on upregulation of VCAM-1 and ICAM-1 expression. We further performed proteomics analysis to identify potential molecular mediators of the IL17A/IL17RD signaling axis. Collectively, our results underscore a critical role for Il17rd in the regulation of aortic myeloid cell infiltration in the context of Western diet feeding.
Collapse
Affiliation(s)
- Shivangi Pande
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA
| | - Calvin Vary
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA
| | - Xuehui Yang
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Lucy Liaw
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA
| | - Lindsey Gower
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Robert Friesel
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA.
| | - Igor Prudovsky
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA.
| | - Sergey Ryzhov
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, 81 Research Drive, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, 04496, USA.
| |
Collapse
|
3
|
Aweida D, Cohen S. The AAA-ATPase ATAD1 and its partners promote degradation of desmin intermediate filaments in muscle. EMBO Rep 2022; 23:e55175. [PMID: 36278411 PMCID: PMC9724657 DOI: 10.15252/embr.202255175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/13/2022] [Accepted: 10/04/2022] [Indexed: 12/12/2022] Open
Abstract
Maintenance of desmin intermediate filaments (IF) is vital for muscle plasticity and function, and their perturbed integrity due to accelerated loss or aggregation causes atrophy and myopathies. Calpain-1-mediated disassembly of ubiquitinated desmin IF is a prerequisite for desmin loss, myofibril breakdown, and atrophy. Because calpain-1 does not harbor a bona fide ubiquitin-binding domain, the precise mechanism for desmin IF disassembly remains unknown. Here, we demonstrate that the AAA-ATPase, ATAD1, is required to facilitate disassembly and turnover of ubiquitinated desmin IF. We identified PLAA and UBXN4 as ATAD1's interacting partners, and their downregulation attenuated desmin loss upon denervation. The ATAD1-PLAA-UBXN4 complex binds desmin filaments and promotes a release of phosphorylated and ubiquitinated species into the cytosol, presenting ATAD1 as the only known AAA-ATPase that preferentially acts on phosphorylated substrates. Desmin filaments disassembly was accelerated by the coordinated functions of Atad1 and calpain-1, which interact in muscle. Thus, by extracting ubiquitinated desmin from the insoluble filament, ATAD1 may expose calpain-1 cleavage sites on desmin, consequently enhancing desmin solubilization and degradation in the cytosol.
Collapse
Affiliation(s)
- Dina Aweida
- Faculty of BiologyTechnion Institute of TechnologyHaifaIsrael
| | - Shenhav Cohen
- Faculty of BiologyTechnion Institute of TechnologyHaifaIsrael
| |
Collapse
|
4
|
Regulation of stability and inhibitory activity of the tumor suppressor SEF through casein-kinase II-mediated phosphorylation. Cell Signal 2021; 86:110085. [PMID: 34280495 DOI: 10.1016/j.cellsig.2021.110085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/23/2022]
Abstract
Inflammation and cancer are intimately linked. A key mediator of inflammation is the transcription-factor NF-κB/RelA:p50. SEF (also known as IL-17RD) is a feedback antagonist of NF-κB/RelA:p50 that is emerging as an important link between inflammation and cancer. SEF acts as a buffer to prevent excessive NF-κB activity by sequestering NF-κB/RelA:p50 in the cytoplasm of unstimulated cells, and consequently attenuating the NF-κB response upon pro-inflammatory cytokine stimulation. SEF contributes to cancer progression also via modulating other signaling pathways, including those triggered by growth-factors. Despite its important role in human physiology and pathology, mechanisms that regulate SEF biochemical properties and inhibitory activity are unknown. Here we show that human SEF is an intrinsically labile protein that is stabilized via CK2-mediated phosphorylation, and identified the residues whom phosphorylation by CK2 stabilizes hSEF. Unlike endogenous SEF, ectopic SEF was rapidly degraded when overexpressed but was stabilized in the presence of excess CK2, suggesting a mechanism for limiting SEF levels depending upon CK2 processivity. Additionally, phosphorylation by CK2 potentiated hSef interaction with NF-κB in cell-free binding assays. Most importantly, we identified a CK2 phosphorylation site that was indispensable for SEF inhibition of pro-inflammatory cytokine signaling but was not required for SEF inhibition of growth-factor signaling. To our knowledge, this is the first demonstration of post-translational modifications that regulate SEF at multiple levels to optimize its inhibitory activity in a specific signaling context. These findings may facilitate the design of SEF variants for treating cytokine-dependent pathologies, including cancer and chronic inflammation.
Collapse
|
5
|
Szybowska P, Kostas M, Wesche J, Haugsten EM, Wiedlocha A. Negative Regulation of FGFR (Fibroblast Growth Factor Receptor) Signaling. Cells 2021; 10:cells10061342. [PMID: 34071546 PMCID: PMC8226934 DOI: 10.3390/cells10061342] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
FGFR (fibroblast growth factor receptor) signaling controls fundamental processes in embryonic, fetal and adult human life. The magnitude, duration, and location of FGFR signaling must be strictly controlled in order to induce the correct biological response. Uncontrolled receptor signaling has been shown to lead to a variety of diseases, such as skeletal disorders and cancer. Here we review the numerous cellular mechanisms that regulate and turn off FGFR signaling, once the receptor is activated. These mechanisms include endocytosis and endocytic sorting, phosphatase activity, negative regulatory proteins and negative feedback phosphorylation events. The mechanisms act together simultaneously or sequentially, controlling the same or different steps in FGFR signaling. Although more work is needed to fully understand the regulation of FGFR signaling, it is clear that the cells in our body have evolved an extensive repertoire of mechanisms that together keep FGFR signaling tightly controlled and prevent excess FGFR signaling.
Collapse
Affiliation(s)
- Patrycja Szybowska
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway; (P.S.); (M.K.); (J.W.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
| | - Michal Kostas
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway; (P.S.); (M.K.); (J.W.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
| | - Jørgen Wesche
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway; (P.S.); (M.K.); (J.W.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
| | - Ellen Margrethe Haugsten
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway; (P.S.); (M.K.); (J.W.)
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
- Correspondence: (E.M.H.); (A.W.); Tel.: +47-2278-1785 (E.M.H.); +47-2278-1930 (A.W.)
| | - Antoni Wiedlocha
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
- Correspondence: (E.M.H.); (A.W.); Tel.: +47-2278-1785 (E.M.H.); +47-2278-1930 (A.W.)
| |
Collapse
|
6
|
Girondel C, Meloche S. Interleukin-17 Receptor D in Physiology, Inflammation and Cancer. Front Oncol 2021; 11:656004. [PMID: 33833999 PMCID: PMC8021910 DOI: 10.3389/fonc.2021.656004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/09/2021] [Indexed: 12/15/2022] Open
Abstract
Interleukin-17 receptor D (IL-17RD) is an evolutionarily conserved member of the IL-17 receptor family. Originally identified as a negative regulator of fibroblast growth factor (FGF) signaling under the name of Sef (Similar expression to FGF genes), IL-17RD was subsequently reported to regulate other receptor tyrosine kinase signaling pathways. In addition, recent studies have shown that IL-17RD also modulates IL-17 and Toll-like receptor (TLR) signaling. Combined genetic and cell biology studies have implicated IL-17RD in the control of cell proliferation and differentiation, cell survival, lineage specification, and inflammation. Accumulating evidence also suggest a role for IL-17RD in tumorigenesis. Expression of IL-17RD is down-regulated in various human cancers and recent work has shown that loss of IL-17RD promotes tumor formation in mice. However, the exact mechanisms underlying the tumor suppressor function of IL-17RD remain unclear and some studies have proposed that IL-17RD may exert pro-tumorigenic effects in certain contexts. Here, we provide an overview of the signaling functions of IL-17RD and review the evidence for its involvement in cancer.
Collapse
Affiliation(s)
- Charlotte Girondel
- Institute for Research in Immunology and Cancer, Montreal, QC, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Montreal, QC, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
7
|
Pande S, Yang X, Friesel R. Interleukin-17 receptor D (Sef) is a multi-functional regulator of cell signaling. Cell Commun Signal 2021; 19:6. [PMID: 33436016 PMCID: PMC7805053 DOI: 10.1186/s12964-020-00695-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022] Open
Abstract
Interleukin-17 receptor D (IL17RD or IL-17RD) also known as Sef (similar expression to fibroblast growth factor), is a single pass transmembrane protein that is reported to regulate several signaling pathways
. IL17RD was initially described as a feedback inhibitor of fibroblast growth factor (FGF) signaling during zebrafish and frog development. It was subsequently determined to regulate other receptor tyrosine kinase signaling cascades as well as several proinflammatory signaling pathways including Interleukin-17A (IL17A), Toll-like receptors (TLR) and Interleukin-1α (IL1α) in several vertebrate species including humans. This review will provide an overview of IL17RD regulation of signaling pathways and functions with emphasis on regulation of development and pathobiological conditions. We will also discuss gaps in our knowledge about IL17RD function to provide insight into opportunities for future investigation. Video Abstract
Collapse
Affiliation(s)
- Shivangi Pande
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04496, USA
| | - Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Robert Friesel
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA. .,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04496, USA.
| |
Collapse
|
8
|
Korsensky L, Haif S, Heller R, Rabinovitz S, Haddad-Halloun J, Dahan N, Ron D. The tumor suppressor Sef is a scaffold for the classical NF-κB/RELA:P50 signaling module. Cell Signal 2019; 59:110-121. [DOI: 10.1016/j.cellsig.2019.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/26/2019] [Accepted: 01/26/2019] [Indexed: 02/07/2023]
|
9
|
Cross-Talk between Fibroblast Growth Factor Receptors and Other Cell Surface Proteins. Cells 2019; 8:cells8050455. [PMID: 31091809 PMCID: PMC6562592 DOI: 10.3390/cells8050455] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/14/2022] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) constitute signaling circuits that transmit signals across the plasma membrane, regulating pivotal cellular processes like differentiation, migration, proliferation, and apoptosis. The malfunction of FGFs/FGFRs signaling axis is observed in numerous developmental and metabolic disorders, and in various tumors. The large diversity of FGFs/FGFRs functions is attributed to a great complexity in the regulation of FGFs/FGFRs-dependent signaling cascades. The function of FGFRs is modulated at several levels, including gene expression, alternative splicing, posttranslational modifications, and protein trafficking. One of the emerging ways to adjust FGFRs activity is through formation of complexes with other integral proteins of the cell membrane. These proteins may act as coreceptors, modulating binding of FGFs to FGFRs and defining specificity of elicited cellular response. FGFRs may interact with other cell surface receptors, like G-protein-coupled receptors (GPCRs) or receptor tyrosine kinases (RTKs). The cross-talk between various receptors modulates the strength and specificity of intracellular signaling and cell fate. At the cell surface FGFRs can assemble into large complexes involving various cell adhesion molecules (CAMs). The interplay between FGFRs and CAMs affects cell–cell interaction and motility and is especially important for development of the central nervous system. This review summarizes current stage of knowledge about the regulation of FGFRs by the plasma membrane-embedded partner proteins and highlights the importance of FGFRs-containing membrane complexes in pathological conditions, including cancer.
Collapse
|
10
|
Delivery of the gene encoding the tumor suppressor Sef into prostate tumors by therapeutic-ultrasound inhibits both tumor angiogenesis and growth. Sci Rep 2017; 7:15060. [PMID: 29118380 PMCID: PMC5678190 DOI: 10.1038/s41598-017-12408-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 08/30/2017] [Indexed: 11/16/2022] Open
Abstract
Carcinomas constitute over 80% of all human cancer types with no effective therapy for metastatic disease. Here, we demonstrate, for the first time, the efficacy of therapeutic-ultrasound (TUS) to deliver a human tumor suppressor gene, hSef-b, to prostate tumors in vivo. Sef is downregulated in various human carcinomas, in a manner correlating with tumor aggressiveness. In vitro, hSef-b inhibited proliferation of TRAMP C2 cells and attenuated activation of ERK/MAPK and the master transcription factor NF-κB in response to FGF and IL-1/TNF, respectively. In vivo, transfection efficiency of a plasmid co-expressing hSef-b/eGFP into TRAMP C2 tumors was 14.7 ± 2.5% following a single TUS application. Repeated TUS treatments with hSef-b plasmid, significantly suppressed prostate tumor growth (60%) through inhibition of cell proliferation (60%), and reduction in blood vessel density (56%). In accordance, repeated TUS-treatments with hSef-b significantly inhibited in vivo expression of FGF2 and MMP-9. FGF2 is a known mitogen, and both FGF2/MMP-9 are proangiogenic factors. Taken together our results strongly suggest that hSef-b acts in a cell autonomous as well as non-cell autonomous manner. Moreover, the study demonstrates the efficacy of non-viral TUS-based hSef-b gene delivery approach for the treatment of prostate cancer tumors, and possibly other carcinomas where Sef is downregulated.
Collapse
|
11
|
Neben CL, Lo M, Jura N, Klein OD. Feedback regulation of RTK signaling in development. Dev Biol 2017; 447:71-89. [PMID: 29079424 DOI: 10.1016/j.ydbio.2017.10.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/17/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023]
Abstract
Precise regulation of the amplitude and duration of receptor tyrosine kinase (RTK) signaling is critical for the execution of cellular programs and behaviors. Understanding these control mechanisms has important implications for the field of developmental biology, and in recent years, the question of how augmentation or attenuation of RTK signaling via feedback loops modulates development has become of increasing interest. RTK feedback regulation is also important for human disease research; for example, germline mutations in genes that encode RTK signaling pathway components cause numerous human congenital syndromes, and somatic alterations contribute to the pathogenesis of diseases such as cancers. In this review, we survey regulators of RTK signaling that tune receptor activity and intracellular transduction cascades, with a focus on the roles of these genes in the developing embryo. We detail the diverse inhibitory mechanisms utilized by negative feedback regulators that, when lost or perturbed, lead to aberrant increases in RTK signaling. We also discuss recent biochemical and genetic insights into positive regulators of RTK signaling and how these proteins function in tandem with negative regulators to guide embryonic development.
Collapse
Affiliation(s)
- Cynthia L Neben
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA
| | - Megan Lo
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco 94143, USA.
| |
Collapse
|
12
|
Moriya N, Shibasaki S, Karasaki M, Iwasaki T. The Impact of MicroRNA-223-3p on IL-17 Receptor D Expression in Synovial Cells. PLoS One 2017; 12:e0169702. [PMID: 28056105 PMCID: PMC5215929 DOI: 10.1371/journal.pone.0169702] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 12/20/2016] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is an autoimmune inflammatory disease affecting joints. Elevated plasma levels of microRNA-223-3p (miR-223-3p) in patients with RA are implicated in the pathogenesis of the disease. This study aimed to analyze the functional role of miR-223-3p in the pathogenesis of RA by overexpressing miR-223-3p in synovial cell lines. METHODS Arthritis was induced in the RA model of SKG mice by injection of ß-glucan. The histopathologic features of joints were examined using hematoxylin and eosin and immunohistochemical staining. Plasma levels of miRNA were determined by panel real-time PCR analysis. Target genes of the differentially expressed miRNAs in SKG mice were analyzed using miRNA target prediction algorithms. The dual-luciferase reporter system was used to evaluate the relationship between miR-223-3p and IL-17 receptor D (IL-17RD). The activity of miR-223-3p was analyzed by transfection of plasmid vectors overexpressing miR-223-3p into IL-17RD-expressing NIH3T3 and MH7A cell lines. Il6 and Il17rd mRNA expression was analyzed by quantitative real-time PCR. IL-17RD protein expression was analyzed by western blot analysis. RESULTS We identified 17 upregulated miRNAs (fold change > 2.0) in plasma of SKG mice injected with ß-glucan relative to untreated SKG mice. Il17rd was identified as the candidate target gene of miR-223-3p using five miRNA target prediction algorithms. The transfection of plasmid vectors overexpressing miR-223-3p into NIH3T3 and MH7A cells resulted in the downregulation of Il17rd expression and upregulation of Il6 expression. Expression of miR-223-3p and Il6 mRNA in MH7A cells was upregulated; however, that of Il17rd mRNA was downregulated following TNF-α stimulation. IL-17RD expression in synovial tissues from SKG mice and RA patients was inversely correlated with the severity of arthritis. CONCLUSION This study is the first to demonstrate that miR-223-3p downregulates IL-17RD in both mouse and human cells; miR-223-3p may contribute to the pathogenesis of RA by downregulating the expression of IL-17RD and upregulating that of IL-6 in synovial cells.
Collapse
Affiliation(s)
- Nozomu Moriya
- Department of Biopharmaceutics, School of Pharmacy, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-Ku, Kobe, Hyogo, Japan
| | - Seiji Shibasaki
- General Education Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo, Japan
| | - Miki Karasaki
- General Education Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo, Japan
| | - Tsuyoshi Iwasaki
- Division of Rheumatology, Department of Internal Medicine, Hyogo College of Medicine, 1-1Mukogawa-cho, Nishinomiya, Hyogo, Japan
- Department of Pharmacotherapy, School of Pharmacy, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo, Japan
- * E-mail:
| |
Collapse
|
13
|
Regulation of FGF signaling: Recent insights from studying positive and negative modulators. Semin Cell Dev Biol 2016; 53:101-14. [DOI: 10.1016/j.semcdb.2016.01.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/19/2016] [Indexed: 11/19/2022]
|
14
|
He Q, Gong Y, Gower L, Yang X, Friesel RE. Sef Regulates Epithelial-Mesenchymal Transition in Breast Cancer Cells. J Cell Biochem 2016; 117:2346-56. [PMID: 26950413 DOI: 10.1002/jcb.25532] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 11/09/2022]
Abstract
Sef (similar expression to fgf), also know as IL17RD, is a transmembrane protein shown to inhibit fibroblast growth factor signaling in developmental and cancer contexts; however, its role as a tumor suppressor remains to be fully elucidated. Here, we show that Sef regulates epithelial-mesenchymal transition (EMT) in breast cancer cell lines. Sef expression was highest in the normal breast epithelial cell line MCF10A, intermediate expression in MCF-7 cells and lowest in MDA-MB-231 cells. Knockdown of Sef increased the expression of genes associated with EMT, and promoted cell migration, invasion, and a fibroblastic morphology of MCF-7 cells. Overexpression of Sef inhibited the expression of EMT marker genes and inhibited cell migration and invasion in MCF-7 cells. Induction of EMT in MCF10A cells by TGF-β and TNF-α resulted in downregulation of Sef expression concomitant with upregulation of EMT gene expression and loss of epithelial morphology. Overexpression of Sef in MCF10A cells partially blocked cytokine-induced EMT. Sef was shown to block β-catenin mediated luciferase reporter activity and to cause a decrease in the nuclear localization of active β-catenin. Furthermore, Sef was shown to co-immunoprecipitate with β-catenin. In a mouse orthotopic xenograft model, Sef overexpression in MDA-MB-231 cells slowed tumor growth and reduced expression of EMT marker genes. Together, these data indicate that Sef plays a role in the negative regulation of EMT in a β-catenin dependent manner and that reduced expression of Sef in breast tumor cells may be permissive for EMT and the acquisition of a more metastatic phenotype. J. Cell. Biochem. 117: 2346-2356, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Qing He
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine.,Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, Maine
| | - Yan Gong
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine.,Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, Maine
| | - Lindsey Gower
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | - Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | - Robert E Friesel
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine.,Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, Maine
| |
Collapse
|
15
|
Orphan receptor IL-17RD regulates Toll-like receptor signalling via SEFIR/TIR interactions. Nat Commun 2015; 6:6669. [PMID: 25808990 DOI: 10.1038/ncomms7669] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 02/18/2015] [Indexed: 11/08/2022] Open
Abstract
Receptor families of the innate immune response engage in 'cross-talk' to tailor optimal immune responses against invading pathogens. However, these responses are subject to multiple levels of regulation to keep in check aberrant inflammatory signals. Here, we describe a role for the orphan receptor interleukin-17 receptor D (IL-17RD) in negatively regulating Toll-like receptor (TLR)-induced responses. Deficiency of IL-17RD expression in cells leads to enhanced pro-inflammatory signalling and gene expression in response to TLR stimulation, and Il17rd(-/-) mice are more susceptible to TLR-induced septic shock. We demonstrate that the intracellular Sef/IL-17R (SEFIR) domain of IL-17RD targets TIR adaptor proteins to inhibit TLR downstream signalling thus revealing a paradigm involving cross-regulation of members of the IL-17R and TLR families.
Collapse
|
16
|
Lutwak E, Price CA, Abramovich SS, Rabinovitz S, Granot I, Dekel N, Ron D. Expression and regulation of the tumor suppressor, SEF, during folliculogenesis in humans and mice. Reproduction 2014; 148:507-17. [PMID: 25118304 DOI: 10.1530/rep-14-0070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Similar expression to FGF (Sef or IL17-RD), is a tumor suppressor and an inhibitor of growth factors as well as of pro-inflammatory cytokine signaling. In this study, we examined the regulation of Sef expression by gonadotropins during ovarian folliculogenesis. In sexually immature mice, in situ hybridization (ISH) localized Sef gene expression to early developing oocytes and granulosa cells (GC) but not to theca cells. Sef was also expressed in mouse ovarian endothelial cells, in the fallopian tube epithelium as well as in adipose tissue venules. SEF protein expression, determined by immunohistochemistry (IHC), correlated well with Sef mRNA expression in GC, while differential expression was noticed in oocytes. High Sef mRNA but undetectable SEF protein levels were observed in the oocytes of primary/secondary follicles, while an inverse correlation was found in the oocytes of preantral and small antral follicles. Sef mRNA expression dropped after pregnant mare's serum gonadotropin (PMSG) administration, peaked at 6-8 h after human chorionic gonadotropin (hCG) treatment, and declined by 12 h after this treatment. ISH and IHC localized the changes to oocytes and mural GC following PMSG treatment, whereas Sef expression increased in mural GC and declined in granulosa-lutein cells upon hCG treatment. The ovarian expression of SEF was confirmed using human samples. ISH localized SEF transcripts to human GC of antral follicles but not to corpora lutea. Furthermore, SEF mRNA was detected in human GC recovered from preovulatory follicles. These results are the first to demonstrate SEF expression in a healthy ovary during folliculogenesis. Hormonal regulation of its expression suggests that SEF may be an important factor involved in intra-ovarian control mechanisms.
Collapse
Affiliation(s)
- Ela Lutwak
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Christopher A Price
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Sagit-Sela Abramovich
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Shiri Rabinovitz
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Irit Granot
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Nava Dekel
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| | - Dina Ron
- Department of BiologyTechnion, Israel Institute of Technology, 32000 Haifa, IsraelFaculté de Médecine VétérinaireCentre de Recherche en Reproduction Animale, Université de Montréal, St-Hyacinthe, Quebec, CanadaDepartment of Biological RegulationThe Weizmann Institute of Science, Rehovot, IsraelIVF UnitDepartment of Obstetrics and Gynecology, Kaplan Medical Center (Affiliated to the Medical School of the Hebrew University and Hadassah, Jerusalem), Rehovot, Israel
| |
Collapse
|
17
|
Peng W, Lei Q, Jiang Z, Hu Z. Characterization of Golgi scaffold proteins and their roles in compartmentalizing cell signaling. J Mol Histol 2013; 45:435-45. [PMID: 24337566 DOI: 10.1007/s10735-013-9560-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 12/02/2013] [Indexed: 12/21/2022]
Abstract
Subcellular compartmentalization has become an important theme in cell signaling. In particular, the Golgi apparatus (GA) plays a prominent role in compartmentalizing signaling cascades that originate at the plasma membrane or other organelles. To precisely regulate this process, cells have evolved a unique class of organizer proteins, termed "scaffold proteins". Sef, PAQR3, PAQR10 and PAQR11 are scaffold proteins that have recently been identified on the GA and are referred to as Golgi scaffolds. The major cell growth signaling pathways, such as Ras/MAPK, PI3K/AKT, insulin and VEGF (vascular endothelial growth factor), are tightly regulated spatially and temporally by these Golgi scaffolds to ensure a physiologically appropriate outcome. Here, we discuss the subcellular localization and characterization of the topology and functional domains of these Golgi scaffolds and summarize their roles in the compartmentalization of cell signaling. We also highlight the physiological and pathological roles of these Golgi scaffolds in tumorigenesis and developmental disorders.
Collapse
Affiliation(s)
- Wenna Peng
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | | | | | | |
Collapse
|
18
|
Abstract
The fibroblast growth factor receptors (FGFRs) regulate important biological processes including cell proliferation and differentiation during development and tissue repair. Over the past decades, numerous pathological conditions and developmental syndromes have emerged as a consequence of deregulation in the FGFRs signaling network. This review aims to provide an overview of FGFR family, their complex signaling pathways in tumorigenesis, and the current development and application of therapeutics targeting the FGFRs signaling for treatment of refractory human cancers.
Collapse
Affiliation(s)
- Kai Hung Tiong
- School of Postgraduate Studies and Research, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Li Yen Mah
- School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, International Medical University, 126 Jalan 19/155B, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Chee-Onn Leong
- School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, International Medical University, 126 Jalan 19/155B, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Sun X, Wang Y, Yang S, Ren F, Xia Y, Chang Z. Activation of TAK1 by Sef-S induces apoptosis in 293T cells. Cell Signal 2013; 25:2039-46. [PMID: 23770285 DOI: 10.1016/j.cellsig.2013.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/04/2013] [Indexed: 11/18/2022]
Abstract
Sef (similar expression to fgf genes, also named IL-17RD) was identified as a negative regulator of fibroblast growth factor signaling. Sef-S, an alternative splice isoform of Sef, inhibits FGF-induced NIH3T3 cell proliferation. Here we report that Sef-S physically interacts with TAK1, induces Lys63-linked TAK1 polyubiquitination on lysine 209 and TAK1-mediated JNK and p38 activation. Co-overexpression of TAK1 WT, K34R, K150R, K158R mutants with Sef-S induces Lys63-linked TAK1 polyubiquitination whereas TAK1 K63R and K209R mutants fail. Furthermore, co-overexpression of Sef-S and TAK1 induce 293T cells apoptosis. These results reveal Sef-S actives Lys63-linked TAK1 polyubiquitination on lysine 209, induces TAK1-mediated JNK and p38 activation and also results apoptosis in 293T cells.
Collapse
Affiliation(s)
- Xiaojun Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China
| | | | | | | | | | | |
Collapse
|
20
|
Fuchs Y, Brunwasser M, Haif S, Haddad J, Shneyer B, Goldshmidt-Tran O, Korsensky L, Abed M, Zisman-Rozen S, Koren L, Carmi Y, Apte R, Yang RB, Orian A, Bejar J, Ron D. Sef is an inhibitor of proinflammatory cytokine signaling, acting by cytoplasmic sequestration of NF-κB. Dev Cell 2013; 23:611-23. [PMID: 22975329 DOI: 10.1016/j.devcel.2012.07.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 06/13/2012] [Accepted: 07/17/2012] [Indexed: 12/11/2022]
Abstract
The NF-κB transcription factor controls diverse biological processes. According to the classical model, NF-κB is retained in the cytoplasm of resting cells via binding to inhibitory, IκB proteins and translocates into the nucleus upon their ligand-induced degradation. Here we reveal that Sef, a known tumor suppressor and inhibitor of growth factor signaling, is a spatial regulator of NF-κB. Sef expression is regulated by the proinflammatory cytokines tumor necrosis factor and interleukin-1, and Sef specifically inhibits "classical" NF-κB (p50:p65) activation by these ligands. Like IκBs, Sef sequesters NF-κB in the cytoplasm of resting cells. However, contrary to IκBs, Sef continues to constrain NF-κB nuclear entry upon ligand stimulation. Accordingly, endogenous Sef knockdown markedly enhances stimulus-induced NF-κB nuclear translocation and consequent activity. This study establishes Sef as a feedback antagonist of proinflammatory cytokines and highlights its potential to regulate the crosstalk between proinflammatory cytokine receptors and receptor tyrosine kinases.
Collapse
Affiliation(s)
- Yaron Fuchs
- Department of Biology, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Murphy T, Hori S, Sewell J, Gnanapragasam VJ. Expression and functional role of negative signalling regulators in tumour development and progression. Int J Cancer 2010; 127:2491-9. [PMID: 20607827 DOI: 10.1002/ijc.25542] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Alterations in intracellular signalling pathways such as the mitogen-activated protein kinases (MAPKs) are key common mechanisms of tumour development and progression. As such, there has been intense research into developing drugs that can inhibit or attenuate intracellular signalling. In recent years, there has been increasing recognition that the cell already has innate negative regulatory proteins that achieve this in normal homeostasis. These regulators provide a feedback inhibitory mechanism that controls the intensity and duration of activated signalling by exogenous stimuli. Members of this group include Raf kinase inhibitor protein 1, the MAPK phosphatases, the SPROUTY and SPRED families and similar expression to FGF. A number of studies have now demonstrated significant alterations in expression of negative regulators in malignant tissue in different cancer types. In functional studies, manipulated expression of these regulators has been shown to significantly influence tumour cell behaviour and phenotype. Here, we summarise the evidence for the functional expression of negative signalling regulators in tumour growth and progression and discuss their potential role as cancer biomarkers and targets for novel drug therapy.
Collapse
Affiliation(s)
- Tania Murphy
- Hutchison MRC Research Centre, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
22
|
Sef is a negative regulator of fiber cell differentiation in the ocular lens. Differentiation 2010; 80:53-67. [PMID: 20542628 DOI: 10.1016/j.diff.2010.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 05/18/2010] [Accepted: 05/20/2010] [Indexed: 02/03/2023]
Abstract
Growth factor signaling, mediated via receptor tyrosine kinases (RTKs), needs to be tightly regulated in many developmental systems to ensure a physiologically appropriate biological outcome. At one level this regulation may involve spatially and temporally ordered patterns of expression of specific RTK signaling antagonists, such as Sef (similar expression to fgfs). Growth factors, notably FGFs, play important roles in development of the vertebrate ocular lens. FGF induces lens cell proliferation and differentiation at progressively higher concentrations and there is compelling evidence that a gradient of FGF signaling in the eye determines lens polarity and growth patterns. We have recently identified the presence of Sef in the lens, with strongest expression in the epithelial cells. Given the important role for FGFs in lens developmental biology, we employed transgenic mouse strategies to determine if Sef could be involved in regulating lens cell behaviour. Over-expressing Sef specifically in the lens of transgenic mice led to impaired lens and eye development that resulted in microphthalmia. Sef inhibited primary lens fiber cell elongation and differentiation, as well as increased apoptosis, consistent with a block in FGFR-mediated signaling during lens morphogenesis. These results are consistent with growth factor antagonists, such as Sef, being important negative regulators of growth factor signaling. Moreover, the lens provides a useful paradigm as to how opposing gradients of a growth factor and its antagonist could work together to determine and stabilise tissue patterning during development and growth.
Collapse
|
23
|
Murphy T, Darby S, Mathers ME, Gnanapragasam VJ. Evidence for distinct alterations in the FGF axis in prostate cancer progression to an aggressive clinical phenotype. J Pathol 2010; 220:452-60. [PMID: 19960500 DOI: 10.1002/path.2657] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multiple fibroblast growth factor (FGF) axis alterations are known to occur in prostate cancer. Here we simultaneously profiled key components of this axis to determine their relevance in disease progression. An optimized immunohistochemistry protocol was used in expression analysis of FGF2, FGF8, FGFR1, FGFR4, and Sef (similar expression to FGF) in a single TMA of prostate cancer. FGF ligands and receptors were overexpressed in cancers compared to benign samples (p < 0.0001), while Sef expression was reduced (p < 0.0001). There was a positive association between higher grades and increased FGFR4 (p = 0.02), FGF2, and FGF8 (p = 0.002 and p < 0.0001). Sef expression was progressively lower with increasing grade (p = 0.005). Clinical stage was positively associated with FGF2, FGF8, and FGFR4 expression (p = 0.005, 0.03, and 0.012) but not with FGFR1 or Sef expression. Only reduced Sef was associated with bone metastasis (p = 0.02) and was also predictive of subsequent metastasis in initially localized tumours (p = 0.004). Down-regulation of Sef and increased FGFR4 were also the only independent variables associated with disease-specific survival (HR 1.73, p = 0.04 and HR 0.56, p = 0.01). In in vitro studies, silencing Sef enhanced the cell response to FGFs (p < 0.001) and substantially mitigated the effectiveness of an FGFR1 inhibitor. Conversely, increased Sef blocked the response to FGFs and had a comparable suppressive effect to the inhibitor. This study demonstrates that increased FGFR4 and reduced Sef may be critical FGF alterations associated with prostate cancer progression. Sef may also have a role in the tumour response to FGFR inhibition and warrants further investigation in this context.
Collapse
Affiliation(s)
- Tania Murphy
- Translational Prostate Cancer Group, Hutchison MRC Research Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | | | | | | |
Collapse
|
24
|
Similar expression to FGF (Sef) inhibits fibroblast growth factor-induced tumourigenic behaviour in prostate cancer cells and is downregulated in aggressive clinical disease. Br J Cancer 2009; 101:1891-9. [PMID: 19888221 PMCID: PMC2788253 DOI: 10.1038/sj.bjc.6605379] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: The fibroblast growth factor (FGF) axis is an important mitogenic stimulus in prostate carcinogenesis. We have previously reported that transcript level of human similar expression to FGF (hSef), a key regulator of this pathway, is downregulated in clinical prostate cancer. In this study we further analysed the role of hSef in prostate cancer. Methods: hSef function was studied in in vitro and in vivo prostate cancer models using stable over-expression clones. Protein expression of hSef was studied in a comprehensive tissue microarray. Results: Stable over-expression of hSef resulted in reduced in vitro cancer cell proliferation, migration and invasive potential. In an in vivo xenograft model, the expression of hSef significantly retarded prostate tumour growth as compared with empty vector (P=0.03) and non-transfected (P=0.0001) controls. Histological examination further showed a less invasive tumour phenotype and reduced numbers of proliferating cells (P=0.0002). In signalling studies, hSef inhibited FGF-induced ERK phosphorylation, migration to the nucleus and activation of a reporter gene. Constitutively active Ras, however, was able to reverse these effects, suggesting that hSef exerts an effect either above or at the level of Ras in prostate cancer cells. In a large tissue microarray, we observed a significant loss of hSef protein in high-grade (P<0.0001) and metastatic (P<0.0001) prostate cancer. Conclusions: Considered together, the role of hSef in attenuating FGF signalling and evidence of downregulation in advanced tumours argue strongly for a tumour suppressor function in human prostate cancer.
Collapse
|
25
|
Cotton LM, O'Bryan MK, Hinton BT. Cellular signaling by fibroblast growth factors (FGFs) and their receptors (FGFRs) in male reproduction. Endocr Rev 2008; 29:193-216. [PMID: 18216218 PMCID: PMC2528845 DOI: 10.1210/er.2007-0028] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 11/29/2007] [Indexed: 12/25/2022]
Abstract
The major function of the reproductive system is to ensure the survival of the species by passing on hereditary traits from one generation to the next. This is accomplished through the production of gametes and the generation of hormones that function in the maturation and regulation of the reproductive system. It is well established that normal development and function of the male reproductive system is mediated by endocrine and paracrine signaling pathways. Fibroblast growth factors (FGFs), their receptors (FGFRs), and signaling cascades have been implicated in a diverse range of cellular processes including: proliferation, apoptosis, cell survival, chemotaxis, cell adhesion, motility, and differentiation. The maintenance and regulation of correct FGF signaling is evident from human and mouse genetic studies which demonstrate that mutations leading to disruption of FGF signaling cause a variety of developmental disorders including dominant skeletal diseases, infertility, and cancer. Over the course of this review, we will provide evidence for differential expression of FGFs/FGFRs in the testis, male germ cells, the epididymis, the seminal vesicle, and the prostate. We will show that this signaling cascade has an important role in sperm development and maturation. Furthermore, we will demonstrate that FGF/FGFR signaling is essential for normal epididymal function and prostate development. To this end, we will provide evidence for the involvement of the FGF signaling system in the regulation and maintenance of the male reproductive system.
Collapse
Affiliation(s)
- Leanne M Cotton
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
26
|
Lee JM, Kim JY, Cho KW, Lee MJ, Cho SW, Kwak S, Cai J, Jung HS. Wnt11/Fgfr1b cross-talk modulates the fate of cells in palate development. Dev Biol 2008; 314:341-50. [PMID: 18191119 DOI: 10.1016/j.ydbio.2007.11.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 11/20/2007] [Accepted: 11/28/2007] [Indexed: 10/22/2022]
|
27
|
Know thy Sef: A novel class of feedback antagonists of receptor tyrosine kinase signaling. Int J Biochem Cell Biol 2008; 40:2040-52. [DOI: 10.1016/j.biocel.2008.03.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 03/06/2008] [Accepted: 03/06/2008] [Indexed: 02/06/2023]
|
28
|
Zisman-Rozen S, Fink D, Ben-Izhak O, Fuchs Y, Brodski A, Kraus MH, Bejar J, Ron D. Downregulation of Sef, an inhibitor of receptor tyrosine kinase signaling, is common to a variety of human carcinomas. Oncogene 2007; 26:6093-8. [PMID: 17420726 DOI: 10.1038/sj.onc.1210424] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Carcinomas are tumors of epithelial origin accounting for over 80% of all human malignancies. A substantial body of evidence implicates oncogenic signaling by receptor tyrosine kinases (RTKs) in carcinoma development. Here we investigated the expression of Sef, a novel inhibitor of RTK signaling, in normal human epithelial tissues and derived malignancies. Human Sef (hSef) was highly expressed in normal epithelial cells of breast, prostate, thyroid gland and the ovarian surface. By comparison, substantial downregulation of hSef expression was observed in the majority of tumors originating from these epithelia. Among 186 primary carcinomas surveyed by RNA in situ hybridization, hSef expression was undetectable in 116 cases including 72/99 (73%) breast, 11/16 (69%) thyroid, 16/31 (52%) prostate and 17/40 (43%) ovarian carcinomas. Moderate reduction of expression was observed in 17/186, and marked reduction in 40/186 tumors. Only 13/186 cases including 12 low-grade and one intermediate grade tumor retained high hSef expression. The association of hSef downregulation and tumor progression was statistically significant (P<0.001). Functionally, ectopic expression of hSef suppressed proliferation of breast carcinoma cells, whereas inhibition of endogenous hSef expression accelerated fibroblast growth factor and epidermal growth factor-dependent proliferation of cervical carcinoma cells. The inhibitory effect of hSef on cell proliferation combined with consistent downregulation in human carcinoma indicates a tumor suppressor-like role for hSef, and implicates loss of hSef expression as a common mechanism in epithelial neoplasia.
Collapse
Affiliation(s)
- S Zisman-Rozen
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Ren Y, Li Z, Rong Z, Cheng L, Li Y, Wang Z, Chang Z. Tyrosine 330 in hSef is critical for the localization and the inhibitory effect on FGF signaling. Biochem Biophys Res Commun 2007; 354:741-6. [PMID: 17266935 DOI: 10.1016/j.bbrc.2007.01.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2007] [Accepted: 01/09/2007] [Indexed: 11/16/2022]
Abstract
Sef (similar expression to fgf genes) was identified as an inhibitor of FGF signaling. The regulation of this inhibitory effect was largely unknown. In this report we demonstrated that tyrosine 330 in hSef protein plays a critical role in the control of the protein localization and thereby in the regulation of Ras/MAPK signaling pathway. We found that the tyrosine 330 is in the form of the YXXcapital EF, Cyrillic signal context and mutation of this residue resulted in preferred plasma membrane localization of hSef. We also observed that both Sef and SefY330F (where tyrosine is substituted by phenylalanine) interacted and co-localized with FGFR in the co-immunoprecipitation assay, and immunostaining assay respectively. We further revealed that the increased amount of Sef localization in the plasma membrane was coupled with the enhanced inhibitory effect on the FGF signaling pathway, indicating that Sef might exert its inhibitory function on the plasma membrane. This paper revealed that tyrosine 330 is critical for the inhibitory function of Sef on FGF signaling.
Collapse
Affiliation(s)
- Yongming Ren
- Department of Biological Sciences and Biotechnology, School of Medicine, Institute of Biomedicine, State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|