51
|
Watson J, Francavilla C. Regulation of FGF10 Signaling in Development and Disease. Front Genet 2018; 9:500. [PMID: 30405705 PMCID: PMC6205963 DOI: 10.3389/fgene.2018.00500] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022] Open
Abstract
Fibroblast Growth Factor 10 (FGF10) is a multifunctional mesenchymal-epithelial signaling growth factor, which is essential for multi-organ development and tissue homeostasis in adults. Furthermore, FGF10 deregulation has been associated with human genetic disorders and certain forms of cancer. Upon binding to FGF receptors with heparan sulfate as co-factor, FGF10 activates several intracellular signaling cascades, resulting in cell proliferation, differentiation, and invasion. FGF10 activity is modulated not only by heparan sulfate proteoglycans in the extracellular matrix, but also by hormones and other soluble factors. Despite more than 20 years of research on FGF10 functions, context-dependent regulation of FGF10 signaling specificity remains poorly understood. Emerging modes of FGF10 signaling regulation will be described, focusing on the role of FGF10 trafficking and sub-cellular localization, heparan sulfate proteoglycans, and miRNAs. Systems biology approaches based on quantitative proteomics will be considered for globally investigating FGF10 signaling specificity. Finally, current gaps in our understanding of FGF10 functions, such as the relative contribution of receptor isoforms to signaling activation, will be discussed in the context of genetic disorders and tumorigenesis.
Collapse
Affiliation(s)
- Joanne Watson
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
52
|
Holmström TH, Moilanen AM, Ikonen T, Björkman ML, Linnanen T, Wohlfahrt G, Karlsson S, Oksala R, Korjamo T, Samajdar S, Rajagopalan S, Chelur S, Narayanan K, Ramachandra RK, Mani J, Nair R, Gowda N, Anthony T, Dhodheri S, Mukherjee S, Ujjinamatada RK, Srinivas N, Ramachandra M, Kallio PJ. ODM-203, a Selective Inhibitor of FGFR and VEGFR, Shows Strong Antitumor Activity, and Induces Antitumor Immunity. Mol Cancer Ther 2018; 18:28-38. [PMID: 30301864 DOI: 10.1158/1535-7163.mct-18-0204] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/20/2018] [Accepted: 10/04/2018] [Indexed: 11/16/2022]
Abstract
Alterations in the gene encoding for the FGFR and upregulation of the VEGFR are found often in cancer, which correlate with disease progression and unfavorable survival. In addition, FGFR and VEGFR signaling synergistically promote tumor angiogenesis, and activation of FGFR signaling has been described as functional compensatory angiogenic signal following development of resistance to VEGFR inhibition. Several selective small-molecule FGFR kinase inhibitors are currently in clinical development. ODM-203 is a novel, selective, and equipotent inhibitor of the FGFR and VEGFR families. In this report we show that ODM-203 inhibits FGFR and VEGFR family kinases selectively and with equal potency in the low nanomolar range (IC50 6-35 nmol/L) in biochemical assays. In cellular assays, ODM-203 inhibits VEGFR-induced tube formation (IC50 33 nmol/L) with similar potency as it inhibits proliferation in FGFR-dependent cell lines (IC50 50-150 nmol/L). In vivo, ODM-203 shows strong antitumor activity in both FGFR-dependent xenograft models and in an angiogenic xenograft model at similar well-tolerated doses. In addition, ODM-203 inhibits metastatic tumor growth in a highly angiogenesis-dependent kidney capsule syngenic model. Interestingly, potent antitumor activity in the subcutaneous syngenic model correlated well with immune modulation in the tumor microenvironment as indicated by marked decrease in the expression of immune check points PD-1 and PD-L1 on CD8 T cells and NK cells, and increased activation of CD8 T cells. In summary, ODM-203 shows equipotent activity for both FGFR and VEGFR kinase families and antitumor activity in both FGFR and angigogenesis models.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jiju Mani
- Aurigene Discovery Technologies Limited, India
| | - Rashmi Nair
- Aurigene Discovery Technologies Limited, India
| | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Ryan MR, Sohl CD, Luo B, Anderson KS. The FGFR1 V561M Gatekeeper Mutation Drives AZD4547 Resistance through STAT3 Activation and EMT. Mol Cancer Res 2018; 17:532-543. [PMID: 30257990 DOI: 10.1158/1541-7786.mcr-18-0429] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/06/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022]
Abstract
FGFR1 has been implicated in numerous cancer types including squamous cell lung cancer, a subset of non-small cell lung cancer with a dismal 5-year survival rate. Small-molecule inhibitors targeting FGFR1 are currently in clinical trials, with AZD4547 being one of the furthest along; however, the development of drug resistance is a major challenge for targeted therapies. A prevalent mechanism of drug resistance in kinases occurs through mutation of the gatekeeper residue, V561M in FGFR1; however, mechanisms underlying V561M resistance to AZD4547 are not fully understood. Here, the cellular consequences of the V561M gatekeeper mutation were characterized, and it was found that although AZD4547 maintains nanomolar affinity for V561M FGFR1, based on in vitro binding assays, cells expressing V561M demonstrate dramatic resistance to AZD4547 driven by increased STAT3 activation downstream of V561M FGFR1. The data reveal that the V561M mutation biases cells toward a more mesenchymal phenotype, including increased levels of proliferation, migration, invasion, and anchorage-independent growth, which was confirmed using CyTOF, a novel single-cell analysis tool. Using shRNA knockdown, loss of STAT3 restored sensitivity of cancer cells expressing V561M FGFR1 to AZD4547. Thus, the data demonstrate that combination therapies including FGFR and STAT3 may overcome V561M FGFR1-driven drug resistance in the clinic. IMPLICATIONS: The V561M FGFR1 gatekeeper mutation leads to devastating drug resistance through activation of STAT3 and the epithelial-mesenchymal transition; this study demonstrates that FGFR1 inhibitor sensitivity can be restored upon STAT3 knockdown.
Collapse
Affiliation(s)
- Molly R Ryan
- Department of Pharmacology, Yale University, New Haven, Connecticut
| | - Christal D Sohl
- Department of Pharmacology, Yale University, New Haven, Connecticut
| | - BeiBei Luo
- Department of Pharmacology, Yale University, New Haven, Connecticut
| | - Karen S Anderson
- Department of Pharmacology, Yale University, New Haven, Connecticut.
| |
Collapse
|
54
|
Boshoff EL, Fletcher EJR, Duty S. Fibroblast growth factor 20 is protective towards dopaminergic neurons in vivo in a paracrine manner. Neuropharmacology 2018; 137:156-163. [PMID: 29698669 PMCID: PMC6063078 DOI: 10.1016/j.neuropharm.2018.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 01/01/2023]
Abstract
Neuroprotective strategies are an unmet medical need for Parkinson's disease. Fibroblast growth factor 20 (FGF20) enhances survival of cultured dopaminergic neurons but little is known about its in vivo potential. We set out to examine whether manipulation of the FGF20 system affected nigrostriatal tract integrity in rats, to identify which fibroblast growth factor receptors (FGFRs) might reside on dopaminergic neurons and to discover the source of endogenous FGF20 in the substantia nigra (SN). Male Sprague Dawley rats were subject to a partial 6-OHDA lesion alongside treatment with exogenous FGF20 or an FGFR antagonist. Behavioural readouts and tyrosine-hydroxylase (TH) immunohistochemistry were used to evaluate nigrostriatal tract integrity. Fluorescent immunohistochemistry was used to examine FGFR subtype expression on TH-positive dopamine neurons and FGF20 cellular localisation within the SN. FGF20 (2.5 μg/day) significantly protected TH-positive cells in the SN and terminals in the striatum, while reducing the development of motor asymmetry at 5, 8 and 11 days post lesion. Conversely, the FGFR antagonist PD173074 (2 mg/kg) significantly worsened both the 6-OHDA lesion and resultant motor asymmetry. Within the SN, TH-positive cells expressed FGFR1, 3 and 4 while FGF20 co-localised with GFAP-positive astrocytes. In conclusion, FGF20 protects dopaminergic neurons in vivo, an action likely mediated through activation of FGFRs1, 3 or 4 found on these neurons. Given FGF20 is localised to astrocytes in the adult SN, endogenous FGF20 provides its protection of dopamine neurons through a paracrine action. Boosting the endogenous FGF20 production might offer potential as a future therapeutic strategy in Parkinson's disease.
Collapse
Affiliation(s)
- Eugene L Boshoff
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | - Edward J R Fletcher
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | - Susan Duty
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
55
|
Zinkle A, Mohammadi M. A threshold model for receptor tyrosine kinase signaling specificity and cell fate determination. F1000Res 2018; 7:F1000 Faculty Rev-872. [PMID: 29983915 PMCID: PMC6013765 DOI: 10.12688/f1000research.14143.1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/18/2018] [Indexed: 11/20/2022] Open
Abstract
Upon ligand engagement, the single-pass transmembrane receptor tyrosine kinases (RTKs) dimerize to transmit qualitatively and quantitatively different intracellular signals that alter the transcriptional landscape and thereby determine the cellular response. The molecular mechanisms underlying these fundamental events are not well understood. Considering recent insights into the structural biology of fibroblast growth factor signaling, we propose a threshold model for RTK signaling specificity in which quantitative differences in the strength/longevity of ligand-induced receptor dimers on the cell surface lead to quantitative differences in the phosphorylation of activation loop (A-loop) tyrosines as well as qualitative differences in the phosphorylation of tyrosines mediating substrate recruitment. In this model, quantitative differences on A-loop tyrosine phosphorylation result in gradations in kinase activation, leading to the generation of intracellular signals of varying amplitude/duration. In contrast, qualitative differences in the pattern of tyrosine phosphorylation on the receptor result in the recruitment/activation of distinct substrates/intracellular pathways. Commensurate with both the dynamics of the intracellular signal and the types of intracellular pathways activated, unique transcriptional signatures are established. Our model provides a framework for engineering clinically useful ligands that can tune receptor dimerization stability so as to bias the cellular transcriptome to achieve a desired cellular output.
Collapse
Affiliation(s)
- Allen Zinkle
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Moosa Mohammadi
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
56
|
Singh R, Karri D, Shen H, Shao J, Dasgupta S, Huang S, Edwards DP, Ittmann MM, O'Malley BW, Yi P. TRAF4-mediated ubiquitination of NGF receptor TrkA regulates prostate cancer metastasis. J Clin Invest 2018; 128:3129-3143. [PMID: 29715200 DOI: 10.1172/jci96060] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 04/27/2018] [Indexed: 12/29/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are important drivers of cancers. In addition to genomic alterations, aberrant activation of WT RTKs plays an important role in driving cancer progression. However, the mechanisms underlying how RTKs drive prostate cancer remain incompletely characterized. Here we show that non-proteolytic ubiquitination of RTK regulates its kinase activity and contributes to RTK-mediated prostate cancer metastasis. TRAF4, an E3 ubiquitin ligase, is highly expressed in metastatic prostate cancer. We demonstrated here that it is a key player in regulating RTK-mediated prostate cancer metastasis. We further identified TrkA, a neurotrophin RTK, as a TRAF4-targeted ubiquitination substrate that promotes cancer cell invasion and found that inhibition of TrkA activity abolished TRAF4-dependent cell invasion. TRAF4 promoted K27- and K29-linked ubiquitination at the TrkA kinase domain and increased its kinase activity. Mutation of TRAF4-targeted ubiquitination sites abolished TrkA tyrosine autophosphorylation and its interaction with downstream proteins. TRAF4 knockdown also suppressed nerve growth factor (NGF) stimulated TrkA downstream p38 MAPK activation and invasion-associated gene expression. Furthermore, elevated TRAF4 levels significantly correlated with increased NGF-stimulated invasion-associated gene expression in prostate cancer patients, indicating that this signaling axis is significantly activated during oncogenesis. Our results revealed a posttranslational modification mechanism contributing to aberrant non-mutated RTK activation in cancer cells.
Collapse
Affiliation(s)
- Ramesh Singh
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Dileep Karri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Hong Shen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Jiangyong Shao
- Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana, USA
| | - Subhamoy Dasgupta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.,Dan L. Duncan Comprehensive Cancer Center and
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Ping Yi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
57
|
Coleman KG, Crews CM. Proteolysis-Targeting Chimeras: Harnessing the Ubiquitin-Proteasome System to Induce Degradation of Specific Target Proteins. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050430] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Craig M. Crews
- Department of Molecular, Cellular, and Developmental Biology; Department of Chemistry; and Department of Pharmacology, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
58
|
Kumar R, Jain AG, Rashid MU, Ali S, Khetpal N, Hussain I, Ahmad S. HGFR and FGR2: Their Roles in Progression and Metastasis of Esophageal Cancer. ROLE OF TYROSINE KINASES IN GASTROINTESTINAL MALIGNANCIES 2018:1-14. [DOI: 10.1007/978-981-13-1486-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
59
|
Loss of peri-Wolffian duct stromal Frs2α expression in mice leads to abnormal ureteric bud induction and vesicoureteral reflux. Pediatr Res 2017; 82:1022-1029. [PMID: 29135976 PMCID: PMC5701656 DOI: 10.1038/pr.2017.175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 06/21/2017] [Indexed: 11/08/2022]
Abstract
UNLABELLED BackgroundFibroblast growth factor receptor 2 (Fgfr2) deletion from murine peri-Wolffian duct stroma (ST) results in aberrant ureteric bud induction, abnormal ureteral insertion into the bladder, and high rates of vesicoureteral reflux (VUR). It is unclear which receptor docking protein(s) is/are responsible for Fgfr2 actions in these tissues. We investigated whether the docking protein, fibroblast receptor substrate 2α (Frs2α), had a role in peri-Wolffian duct ST similar to Fgfr2.MethodsWe conditionally deleted Frs2α in peri-Wolffian duct ST with a Tbx18cre mouse line (Frs2αST-/-). We assessed for ureteric induction defects and alterations in downstream targets mediating defects. We performed euthanized cystograms and assessed ureter-bladder junctions by three-dimensional (3D) reconstructions.ResultsEmbryonic day (E) 11.5 Frs2αST-/- embryos had many displaced ureteric bud induction sites when compared with controls. E11.0 Frs2αST-/- embryos had decreased Bmp4 expression and signaling, which can cause abnormal ureteric bud induction. Postnatal day 1 (P1) and P30 Frs2αST-/- mice had higher VUR rates and grades vs. CONTROLS Mutant refluxing ureters that inserted improperly into the bladder had shortened intravesicular tunnels (IVTs) when compared with controlsConclusionFrs2αST-/- embryos have aberrant ureteric induction sites, improper ureteral insertion, shortened intravesicular lengths, and VUR. Induction site defects appear secondary to reduced Bmp4 expression, similar to Fgfr2 mutants.
Collapse
|
60
|
Abstract
Mammalian inner ear comprises of six sensory organs; cochlea, utricle, saccule, and three semicircular canals. The cochlea contains sensory epithelium known as the organ of Corti which senses sound through mechanosensory hair cells. Mammalian inner ear undergoes series of morphogenesis during development beginning thickening of ectoderm nearby hindbrain. These events require tight regulation of multiple signaling cascades including FGF, Wnt, Notch and Bmp signaling. In this review, we will discuss the role of newly emerging signaling, FGF signaling, for its roles required for cochlear development. [BMB Reports 2017; 50(10): 487-495].
Collapse
Affiliation(s)
- Michael Ebeid
- Department of Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska,
USA
| | - Sung-Ho Huh
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska,
USA
- Department of Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska,
USA
| |
Collapse
|
61
|
Lei H, Deng CX. Fibroblast Growth Factor Receptor 2 Signaling in Breast Cancer. Int J Biol Sci 2017; 13:1163-1171. [PMID: 29104507 PMCID: PMC5666331 DOI: 10.7150/ijbs.20792] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 05/18/2017] [Indexed: 01/03/2023] Open
Abstract
Fibroblast growth factor receptor 2 (FGFR2) is a membrane-spanning tyrosine kinase that mediates signaling for FGFs. Recent studies detected various point mutations of FGFR2 in multiple types of cancers, including breast cancer, lung cancer, gastric cancer, uterine cancer and ovarian cancer, yet the casual relationship between these mutations and tumorigenesis is unclear. Here we will discuss possible interactions between FGFR2 signaling and several major pathways through which the aberrantly activated FGFR2 signaling may result in breast cancer development. We will also discuss some recent developments in the discovery and application of therapies and strategies for breast cancers by inhibiting FGFR2 activities.
Collapse
Affiliation(s)
- Haipeng Lei
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
62
|
Yang CH, Yeh YJ, Wang JY, Liu YW, Chen YL, Cheng HW, Cheng CM, Chuang YJ, Yuh CH, Chen YR. NEAP/DUSP26 suppresses receptor tyrosine kinases and regulates neuronal development in zebrafish. Sci Rep 2017; 7:5241. [PMID: 28701747 PMCID: PMC5507855 DOI: 10.1038/s41598-017-05584-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022] Open
Abstract
Expression of neuroendocrine-associated phosphatase (NEAP, also named as dual specificity phosphatase 26, [DUSP26]) is restricted to neuroendocrine tissues. We found that NEAP, but not its phosphatase-defective mutant, suppressed nerve growth factor (NGF) receptor TrkA and fibroblast growth factor receptor 1 (FGFR1) activation in PC12 cells upon NGF stimulation. Conversely, suppressing NEAP expression by RNA interference enhanced TrkA and FGFR1 phosphorylation. NEAP was capable of de-phosphorylating TrkA and FGFR1 directly in vitro. NEAP-orthologous gene existed in zebrafish. Morpholino (MO) suppression of NEAP in zebrafish resulted in hyper-phosphorylation of TrkA and FGFR1 as well as abnormal body postures and small eyes. Differentiation of retina in zebrafishes with NEAP MO treatment was severely defective, so were cranial motor neurons. Taken together, our data indicated that NEAP/DUSP26 have a critical role in regulating TrkA and FGFR1 signaling as well as proper development of retina and neuronal system in zebrafish.
Collapse
Affiliation(s)
- Chi-Hwa Yang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Yu-Jung Yeh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Jiz-Yuh Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Yen-Lin Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Hui-Wen Cheng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Chun-Mei Cheng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Yung-Jen Chuang
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Yi-Rong Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 350, Taiwan.
| |
Collapse
|
63
|
Lin X, Song L, He D, Zeng X, Wu J, Luo W, Yang Q, Wang J, Wang T, Cai J, Lin Y, Lai F, Peng W, Wu X. An FGF8b-mimicking peptide with potent antiangiogenic activity. Mol Med Rep 2017; 16:894-900. [PMID: 28560418 DOI: 10.3892/mmr.2017.6651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 01/18/2017] [Indexed: 11/06/2022] Open
Abstract
Fibroblast growth factor (FGF) 8b interacts with its receptors and promotes angiogenesis in hormone‑dependent tumors. In the present study, we demonstrated that a short peptide, termed 8b‑13, which mimics part of the FGF8b structure, significantly inhibited the proliferation and migration of human umbilical vein endothelial cells (HUVECs) triggered by FGF8b using 3‑(4,5‑dimethylthiazol‑2‑yl)‑2,5‑diphenyltetrazolium bromide (MTT), flow cytometry and an in vitro scratch assay. In addition, the findings from western blotting and reverse transcription‑quantitative polymerase chain reaction revealed that 8b‑13 appeared to counteract the effects of FGF8b on the expression of cyclin D1, the activation of signaling cascades, and the expression of proangiogenic factors; these actions may be involved in the mechanism underlying the inhibitory effects of 8b‑13 on FGF8b‑induced HUVEC proliferation and migration. The present results suggested that 8b‑13 may be considered a potent FGF8b antagonist with antiangiogenic activity, and may have potential as a novel therapeutic agent for the treatment of cancer characterized by abnormal FGF8b upregulation.
Collapse
Affiliation(s)
- Xiaomian Lin
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Li Song
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Dan He
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xiangfeng Zeng
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jianzhang Wu
- Department of Pharmacy, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Wu Luo
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Qi Yang
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jizhong Wang
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Tianxiang Wang
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jialong Cai
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Yanling Lin
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Fubin Lai
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Wentao Peng
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xiaoping Wu
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| |
Collapse
|
64
|
FGF-Dependent, Context-Driven Role for FRS Adapters in the Early Telencephalon. J Neurosci 2017; 37:5690-5698. [PMID: 28483978 DOI: 10.1523/jneurosci.2931-16.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 04/07/2017] [Accepted: 04/25/2017] [Indexed: 01/20/2023] Open
Abstract
FGF signaling, an important component of intercellular communication, is required in many tissues throughout development to promote diverse cellular processes. Whether FGF receptors (FGFRs) accomplish such varied tasks in part by activating different intracellular transducers in different contexts remains unclear. Here, we used the developing mouse telencephalon as an example to study the role of the FRS adapters FRS2 and FRS3 in mediating the functions of FGFRs. Using tissue-specific and germline mutants, we examined the requirement of Frs genes in two FGFR-dependent processes. We found that Frs2 and Frs3 are together required for the differentiation of a subset of medial ganglionic eminence (MGE)-derived neurons, but are dispensable for the survival of early telencephalic precursor cells, in which any one of three FGFRs (FGFR1, FGFR2, or FGFR3) is sufficient for survival. Although FRS adapters are dispensable for ERK-1/2 activation, they are required for AKT activation within the subventricular zone of the developing MGE. Using an FRS2,3-binding site mutant of Fgfr1, we established that FRS adapters are necessary for mediating most or all FGFR1 signaling, not only in MGE differentiation, but also in cell survival, implying that other adapters mediate at least in part the signaling from FGFR2 and FGFR3. Our study provides an example of a contextual role for an intracellular transducer and contributes to our understanding of how FGF signaling plays diverse developmental roles.SIGNIFICANCE STATEMENT FGFs promote a range of developmental processes in many developing tissues and at multiple developmental stages. The mechanisms underlying this multifunctionality remain poorly defined in vivo Using telencephalon development as an example, we show here that FRS adapters exhibit some selectivity in their requirement for mediating FGF receptor (FGFR) signaling and activating downstream mediators that depend on the developmental process, with a requirement in neuronal differentiation but not cell survival. Differential engagement of FRS and non-FRS intracellular adapters downstream of FGFRs could therefore in principle explain how FGFs play several distinct roles in other developing tissues and developmental stages.
Collapse
|
65
|
The Function of FGFR1 Signalling in the Spinal Cord: Therapeutic Approaches Using FGFR1 Ligands after Spinal Cord Injury. Neural Plast 2017; 2017:2740768. [PMID: 28197342 PMCID: PMC5286530 DOI: 10.1155/2017/2740768] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 12/25/2016] [Indexed: 11/24/2022] Open
Abstract
Extensive research is ongoing that concentrates on finding therapies to enhance CNS regeneration after spinal cord injury (SCI) and to cure paralysis. This review sheds light on the role of the FGFR pathway in the injured spinal cord and discusses various therapies that use FGFR activating ligands to promote regeneration after SCI. We discuss studies that use peripheral nerve grafts or Schwann cell grafts in combination with FGF1 or FGF2 supplementation. Most of these studies show evidence that these therapies successfully enhance axon regeneration into the graft. Further they provide evidence for partial recovery of sensory function shown by electrophysiology and motor activity evidenced by behavioural data. We also present one study that indicates that combination with additional, synergistic factors might further drive the system towards functional regeneration. In essence, this review summarises the potential of nerve and cell grafts combined with FGF1/2 supplementation to improve outcome even after severe spinal cord injury.
Collapse
|
66
|
Ke K, Li H, Yao H, Shi XN, Dong C, Zhu Y, Liu X, Li L, Leung KS, Wong MH, Liu XD, Kung HF, Lin MCM. In silico prediction and in vitro and in vivo validation of acaricide fluazuron as a potential inhibitor of FGFR3 and a candidate anticancer drug for bladder carcinoma. Chem Biol Drug Des 2016; 89:505-513. [PMID: 27664399 DOI: 10.1111/cbdd.12872] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/31/2016] [Accepted: 09/18/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Kunbin Ke
- Biomedical Engineering Research Center; Kunming Medical University; Kunming Yunnan China
- Department of Urology; The 1st Affiliated Hospital of Kunming Medical University; Kunming China
| | - Hongjian Li
- Institute of Future Cities; Chinese University of Hong Kong; Hong Kong China
- Department of Computer Science and Engineering; Chinese University of Hong Kong; Hong Kong China
| | - Hong Yao
- The Cancer Biotherapy Institute of Jiangsu Province; Xuzhou Medical College; Xuzhou China
| | - Xi-Nan Shi
- Biomedical Engineering Research Center; Kunming Medical University; Kunming Yunnan China
- Department of Pathophysiology; School of Basic Medical Sciences; Yunnan University of TCM; Kunming China
| | - Chao Dong
- Biomedical Engineering Research Center; Kunming Medical University; Kunming Yunnan China
- Department of the Second Medical Oncology; The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital; Kunming China
| | - Ying Zhu
- Biomedical Engineering Research Center; Kunming Medical University; Kunming Yunnan China
- Department of Cadre Medical Branch; The 3rd Affiliated Hospital of Kunming Medical University; Kunming China
| | - Xu Liu
- Biomedical Engineering Research Center; Kunming Medical University; Kunming Yunnan China
| | - Ling Li
- Biomedical Engineering Research Center; Kunming Medical University; Kunming Yunnan China
| | - Kwong-Sak Leung
- Department of Computer Science and Engineering; Chinese University of Hong Kong; Hong Kong China
| | - Man-Hon Wong
- Department of Computer Science and Engineering; Chinese University of Hong Kong; Hong Kong China
| | - Xiao-Dong Liu
- Department of Urology; The 1st Affiliated Hospital of Kunming Medical University; Kunming China
| | - Hsiang-fu Kung
- Biomedical Engineering Research Center; Kunming Medical University; Kunming Yunnan China
- School of Biomedical Sciences; Chinese University of Hong Kong; Hong Kong China
| | - Marie Chia-mi Lin
- Shenzhen Key Lab of Translational Medicine of Tumor; School of Medicine; Shenzhen University; Shenzhen China
| |
Collapse
|
67
|
Chen PY, Qin L, Li G, Tellides G, Simons M. Fibroblast growth factor (FGF) signaling regulates transforming growth factor beta (TGFβ)-dependent smooth muscle cell phenotype modulation. Sci Rep 2016; 6:33407. [PMID: 27634335 PMCID: PMC5025753 DOI: 10.1038/srep33407] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022] Open
Abstract
Smooth muscle cells (SMCs) in normal blood vessels exist in a highly differentiate state characterized by expression of SMC-specific contractile proteins ("contractile phenotype"). Following blood vessel injury in vivo or when cultured in vitro in the presence of multiple growth factors, SMC undergo a phenotype switch characterized by the loss of contractile markers and appearance of expression of non-muscle proteins ("proliferative phenotype"). While a number of factors have been reported to modulate this process, its regulation remains uncertain. Here we show that induction of SMC FGF signaling inhibits TGFβ signaling and converts contractile SMCs to the proliferative phenotype. Conversely, inhibition of SMC FGF signaling induces TGFβ signaling converting proliferating SMCs to the contractile phenotype, even in the presence of various growth factors in vitro or vascular injury in vivo. The importance of this signaling cross-talk is supported by in vivo data that show that an SMC deletion of a pan-FGF receptor adaptor Frs2α (fibroblast growth factor receptor substrate 2 alpha) in mice profoundly reduces neointima formation and vascular remodelling following carotid artery ligation. These results demonstrate that FGF-TGFβ signaling antagonism is the primary regulator of the SMC phenotype switch. Manipulation of this cross-talk may be an effective strategy for treatment of SMC-proliferation related diseases.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Guangxin Li
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Vascular Surgery, The First Hospital of China Medical University, 155 Nanjing Bei Street, Shenyang, China
| | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
68
|
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: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/19/2016] [Indexed: 11/19/2022]
|
69
|
Abstract
The fibroblast growth factor (Fgf) family of ligands and receptor tyrosine kinases is required throughout embryonic and postnatal development and also regulates multiple homeostatic functions in the adult. Aberrant Fgf signaling causes many congenital disorders and underlies multiple forms of cancer. Understanding the mechanisms that govern Fgf signaling is therefore important to appreciate many aspects of Fgf biology and disease. Here we review the mechanisms of Fgf signaling by focusing on genetic strategies that enable in vivo analysis. These studies support an important role for Erk1/2 as a mediator of Fgf signaling in many biological processes but have also provided strong evidence for additional signaling pathways in transmitting Fgf signaling in vivo.
Collapse
Affiliation(s)
- J Richard Brewer
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Pierre Mazot
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| |
Collapse
|
70
|
Haenzi B, Gers-Barlag K, Akhoundzadeh H, Hutson TH, Menezes SC, Bunge MB, Moon LDF. Overexpression of the Fibroblast Growth Factor Receptor 1 (FGFR1) in a Model of Spinal Cord Injury in Rats. PLoS One 2016; 11:e0150541. [PMID: 27015635 PMCID: PMC4807820 DOI: 10.1371/journal.pone.0150541] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/15/2016] [Indexed: 01/03/2023] Open
Abstract
Spinal cord injury (SCI) is a severe condition that affects many people and results in high health care costs. Therefore, it is essential to find new targets for treatment. The fibroblast growth factor receptor 1 (FGFR1) signalling pathway has a history of being explored for SCI treatment. Several groups have examined the effect of high availability of different FGFR1 ligands at the injury site and reported corticospinal tract (CST) regeneration as well as improved motor functions. In this study, we investigated overexpression of the FGFR1 in rat corticospinal neurons in vivo after injury (unilateral pyramidotomy) and in cerebellar granule neurons (CGNs) in vitro. We show that overexpression of FGFR1 using AAV1 intracortical injections did not increase sprouting of the treated corticospinal tract and did not improve dexterity or walking in a rat model of SCI. Furthermore, we show that overexpression of FGFR1 in vitro resulted in decreased neurite outgrowth compared to control. Thus, our results suggest that the FGFR1 is not a suitable therapeutic target after SCI.
Collapse
Affiliation(s)
- Barbara Haenzi
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King’s College London, London, SE1 1UL, United Kingdom
- * E-mail:
| | - Katharina Gers-Barlag
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King’s College London, London, SE1 1UL, United Kingdom
| | - Halima Akhoundzadeh
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King’s College London, London, SE1 1UL, United Kingdom
| | - Thomas H. Hutson
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King’s College London, London, SE1 1UL, United Kingdom
| | - Sean C. Menezes
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King’s College London, London, SE1 1UL, United Kingdom
| | - Mary Bartlett Bunge
- Miami Project to Cure Paralysis, Departments of Cell Biology, Neurological Surgery and Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States of America
| | - Lawrence D. F. Moon
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King’s College London, London, SE1 1UL, United Kingdom
| |
Collapse
|
71
|
Yang X, Liaw L, Prudovsky I, Brooks PC, Vary C, Oxburgh L, Friesel R. Fibroblast growth factor signaling in the vasculature. Curr Atheroscler Rep 2015; 17:509. [PMID: 25813213 DOI: 10.1007/s11883-015-0509-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite their discovery as angiogenic factors and mitogens for endothelial cells more than 30 years ago, much remains to be determined about the role of fibroblast growth factors (FGFs) and their receptors in vascular development, homeostasis, and disease. In vitro studies show that members of the FGF family stimulate growth, migration, and sprouting of endothelial cells, and growth, migration, and phenotypic plasticity of vascular smooth muscle cells. Recent studies have revealed important roles for FGFs and their receptors in the regulation of endothelial cell sprouting and vascular homeostasis in vivo. Furthermore, recent work has revealed roles for FGFs in atherosclerosis, vascular calcification, and vascular dysfunction. The large number of FGFs and their receptors expressed in endothelial and vascular smooth muscle cells complicates these studies. In this review, we summarize recent studies in which new and unanticipated roles for FGFs and their receptors in the vasculature have been revealed.
Collapse
Affiliation(s)
- Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | | | | | | | | | | | | |
Collapse
|
72
|
Brewer JR, Molotkov A, Mazot P, Hoch RV, Soriano P. Fgfr1 regulates development through the combinatorial use of signaling proteins. Genes Dev 2015; 29:1863-74. [PMID: 26341559 PMCID: PMC4573858 DOI: 10.1101/gad.264994.115] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Brewer et al. engineered an allelic series of knock-in point mutations designed to disrupt Fgfr1 signaling functions individually and in combination. They found that, in addition to Frs2, Crk proteins and Plcγ also contribute to Erk1/2 activation. Disruption of all known signaling functions diminished Erk1/2 and Plcγ activation but did not recapitulate the peri-implantation Fgfr1-null phenotype. Fibroblast growth factor (Fgf) signaling governs multiple processes important in development and disease. Many lines of evidence have implicated Erk1/2 signaling induced through Frs2 as the predominant effector pathway downstream from Fgf receptors (Fgfrs), but these receptors can also signal through other mechanisms. To explore the functional significance of the full range of signaling downstream from Fgfrs in mice, we engineered an allelic series of knock-in point mutations designed to disrupt Fgfr1 signaling functions individually and in combination. Analysis of each mutant indicates that Frs2 binding to Fgfr1 has the most pleiotropic functions in development but also that the receptor uses multiple proteins additively in vivo. In addition to Frs2, Crk proteins and Plcγ also contribute to Erk1/2 activation, affecting axis elongation and craniofacial and limb development and providing a biochemical mechanism for additive signaling requirements. Disruption of all known signaling functions diminished Erk1/2 and Plcγ activation but did not recapitulate the peri-implantation Fgfr1-null phenotype. This suggests that Erk1/2-independent signaling pathways are functionally important for Fgf signaling in vivo.
Collapse
Affiliation(s)
- J Richard Brewer
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Andrei Molotkov
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Pierre Mazot
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Renée V Hoch
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA; Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| |
Collapse
|
73
|
Zhang T, Ding Y, An H, Feng L, Wang S. Screening anti-tumor compounds from Ligusticum wallichii
using cell membrane chromatography combined with high-performance liquid chromatography and mass spectrometry. J Sep Sci 2015; 38:3247-3253. [PMID: 26178081 DOI: 10.1002/jssc.201500488] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/25/2015] [Accepted: 07/01/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Tao Zhang
- School of Medicine; Xi'an Jiaotong University; Xi'an China
| | - Yuanyuan Ding
- School of Medicine; Xi'an Jiaotong University; Xi'an China
| | - Hongli An
- Center for Translational Medicine; The First Affiliated Hospital; Xian Jiaotong University College of Medicine; Xi'an China
| | - Liuxin Feng
- Medical Laboratory; Second Affiliated Hospital; School of Medicine, Xi'an Jiaotong University; Xi'an China
| | - Sicen Wang
- School of Medicine; Xi'an Jiaotong University; Xi'an China
| |
Collapse
|
74
|
Frantz LAF, Schraiber JG, Madsen O, Megens HJ, Cagan A, Bosse M, Paudel Y, Crooijmans RPMA, Larson G, Groenen MAM. Evidence of long-term gene flow and selection during domestication from analyses of Eurasian wild and domestic pig genomes. Nat Genet 2015; 47:1141-8. [PMID: 26323058 DOI: 10.1038/ng.3394] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 08/10/2015] [Indexed: 12/18/2022]
Abstract
Traditionally, the process of domestication is assumed to be initiated by humans, involve few individuals and rely on reproductive isolation between wild and domestic forms. We analyzed pig domestication using over 100 genome sequences and tested whether pig domestication followed a traditional linear model or a more complex, reticulate model. We found that the assumptions of traditional models, such as reproductive isolation and strong domestication bottlenecks, are incompatible with the genetic data. In addition, our results show that, despite gene flow, the genomes of domestic pigs have strong signatures of selection at loci that affect behavior and morphology. We argue that recurrent selection for domestic traits likely counteracted the homogenizing effect of gene flow from wild boars and created 'islands of domestication' in the genome. Our results have major ramifications for the understanding of animal domestication and suggest that future studies should employ models that do not assume reproductive isolation.
Collapse
Affiliation(s)
- Laurent A F Frantz
- Animal Breeding and Genomics Group, Wageningen University, Wageningen, the Netherlands.,Palaeogenomics and Bio-Archaeology Research Network, Research Laboratory for Archaeology and History of Art, University of Oxford, Oxford, UK
| | - Joshua G Schraiber
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California, USA.,Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Ole Madsen
- Animal Breeding and Genomics Group, Wageningen University, Wageningen, the Netherlands
| | - Hendrik-Jan Megens
- Animal Breeding and Genomics Group, Wageningen University, Wageningen, the Netherlands
| | - Alex Cagan
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Mirte Bosse
- Animal Breeding and Genomics Group, Wageningen University, Wageningen, the Netherlands
| | - Yogesh Paudel
- Animal Breeding and Genomics Group, Wageningen University, Wageningen, the Netherlands
| | | | - Greger Larson
- Palaeogenomics and Bio-Archaeology Research Network, Research Laboratory for Archaeology and History of Art, University of Oxford, Oxford, UK
| | - Martien A M Groenen
- Animal Breeding and Genomics Group, Wageningen University, Wageningen, the Netherlands
| |
Collapse
|
75
|
Ahmed Z, Timsah Z, Suen KM, Cook NP, Lee GR, Lin CC, Gagea M, Marti AA, Ladbury JE. Grb2 monomer-dimer equilibrium determines normal versus oncogenic function. Nat Commun 2015; 6:7354. [PMID: 26103942 PMCID: PMC4491180 DOI: 10.1038/ncomms8354] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/29/2015] [Indexed: 01/09/2023] Open
Abstract
The adaptor protein growth factor receptor-bound protein 2 (Grb2) is ubiquitously expressed in eukaryotic cells and involved in a multitude of intracellular protein interactions. Grb2 plays a pivotal role in tyrosine kinase-mediated signal transduction including linking receptor tyrosine kinases to the Ras/mitogen-activated protein (MAP) kinase pathway, which is implicated in oncogenic outcome. Grb2 exists in a constitutive equilibrium between monomeric and dimeric states. Here we show that only monomeric Grb2 is capable of binding to SOS and upregulating MAP kinase signalling and that the dimeric state is inhibitory to this process. Phosphorylation of tyrosine 160 (Y160) on Grb2, or binding of a tyrosylphosphate-containing ligand to the SH2 domain of Grb2, results in dimer dissociation. Phosphorylation of Y160 on Grb2 is readily detectable in the malignant forms of human prostate, colon and breast cancers. The self-association/dissociation of Grb2 represents a switch that regulates MAP kinase activity and hence controls cancer progression.
Collapse
Affiliation(s)
- Zamal Ahmed
- 1] Department of Biochemistry and Molecular Biology, University of Texas, M.D. Anderson Cancer Center, Unit 1000, 1515 Holcombe Boulevard, Houston, Texas 77030, USA [2] Center for Biomolecular Structure and Function, University of Texas, M.D. Anderson Cancer Center, Unit 1000, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Zahra Timsah
- 1] Department of Biochemistry and Molecular Biology, University of Texas, M.D. Anderson Cancer Center, Unit 1000, 1515 Holcombe Boulevard, Houston, Texas 77030, USA [2] School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Kin M Suen
- 1] Department of Biochemistry and Molecular Biology, University of Texas, M.D. Anderson Cancer Center, Unit 1000, 1515 Holcombe Boulevard, Houston, Texas 77030, USA [2] School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Nathan P Cook
- Department of Chemistry, Rice University, Houston, Texas 77005, USA
| | - Gilbert R Lee
- Center for Biomolecular Structure and Function, University of Texas, M.D. Anderson Cancer Center, Unit 1000, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Chi-Chuan Lin
- 1] Department of Biochemistry and Molecular Biology, University of Texas, M.D. Anderson Cancer Center, Unit 1000, 1515 Holcombe Boulevard, Houston, Texas 77030, USA [2] School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, University of Texas, M.D. Anderson Cancer Center, Unit 63, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Angel A Marti
- Department of Chemistry, Rice University, Houston, Texas 77005, USA
| | - John E Ladbury
- 1] Department of Biochemistry and Molecular Biology, University of Texas, M.D. Anderson Cancer Center, Unit 1000, 1515 Holcombe Boulevard, Houston, Texas 77030, USA [2] Center for Biomolecular Structure and Function, University of Texas, M.D. Anderson Cancer Center, Unit 1000, 1515 Holcombe Boulevard, Houston, Texas 77030, USA [3] School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
76
|
Zeng L, Kuti M, Mujtaba S, Zhou MM. Structural insights into FRS2α PTB domain recognition by neurotrophin receptor TrkB. Proteins 2015; 82:1534-41. [PMID: 24470253 DOI: 10.1002/prot.24523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/27/2013] [Accepted: 01/16/2014] [Indexed: 11/07/2022]
Abstract
The fibroblast growth factor receptor (FGFR) substrate 2 (FRS2) family proteins function as scaffolding adapters for receptor tyrosine kinases (RTKs). The FRS2α proteins interact with RTKs through the phosphotyrosine-binding (PTB) domain and transfer signals from the activated receptors to downstream effector proteins. Here, we report the nuclear magnetic resonance structure of the FRS2α PTB domain bound to phosphorylated TrkB. The structure reveals that the FRS2α-PTB domain is comprised of two distinct but adjacent pockets for its mutually exclusive interaction with either nonphosphorylated juxtamembrane region of the FGFR, or tyrosine phosphorylated peptides TrkA and TrkB. The new structural insights suggest rational design of selective small molecules through targeting of the two conjunct pockets in the FRS2α PTB domain.
Collapse
Affiliation(s)
- Lei Zeng
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029
| | | | | | | |
Collapse
|
77
|
Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:215-66. [PMID: 25772309 PMCID: PMC4393358 DOI: 10.1002/wdev.176] [Citation(s) in RCA: 1383] [Impact Index Per Article: 138.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
The signaling component of the mammalian Fibroblast Growth Factor (FGF) family is comprised of eighteen secreted proteins that interact with four signaling tyrosine kinase FGF receptors (FGFRs). Interaction of FGF ligands with their signaling receptors is regulated by protein or proteoglycan cofactors and by extracellular binding proteins. Activated FGFRs phosphorylate specific tyrosine residues that mediate interaction with cytosolic adaptor proteins and the RAS-MAPK, PI3K-AKT, PLCγ, and STAT intracellular signaling pathways. Four structurally related intracellular non-signaling FGFs interact with and regulate the family of voltage gated sodium channels. Members of the FGF family function in the earliest stages of embryonic development and during organogenesis to maintain progenitor cells and mediate their growth, differentiation, survival, and patterning. FGFs also have roles in adult tissues where they mediate metabolic functions, tissue repair, and regeneration, often by reactivating developmental signaling pathways. Consistent with the presence of FGFs in almost all tissues and organs, aberrant activity of the pathway is associated with developmental defects that disrupt organogenesis, impair the response to injury, and result in metabolic disorders, and cancer. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of MedicineSt. Louis, MO, USA
- *
Correspondence to:
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto UniversitySakyo, Kyoto, Japan
| |
Collapse
|
78
|
Tremblay MG, Herdman C, Guillou F, Mishra PK, Baril J, Bellenfant S, Moss T. Extended Synaptotagmin Interaction with the Fibroblast Growth Factor Receptor Depends on Receptor Conformation, Not Catalytic Activity. J Biol Chem 2015; 290:16142-56. [PMID: 25922075 DOI: 10.1074/jbc.m115.656918] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Indexed: 11/06/2022] Open
Abstract
We previously demonstrated that ESyt2 interacts specifically with the activated FGF receptor and is required for a rapid phase of receptor internalization and for functional signaling via the ERK pathway in early Xenopus embryos. ESyt2 is one of the three-member family of Extended Synaptotagmins that were recently shown to be implicated in the formation of endoplasmic reticulum (ER)-plasma membrane (PM) junctions and in the Ca(2+) dependent regulation of these junctions. Here we show that ESyt2 is directed to the ER by its putative transmembrane domain, that the ESyts hetero- and homodimerize, and that ESyt2 homodimerization in vivo requires a TM adjacent sequence but not the SMP domain. ESyt2 and ESyt3, but not ESyt1, selectively interact in vivo with activated FGFR1. In the case of ESyt2, this interaction requires a short TM adjacent sequence and is independent of receptor autophosphorylation, but dependent on receptor conformation. The data show that ESyt2 recognizes a site in the upper kinase lobe of FGFR1 that is revealed by displacement of the kinase domain activation loop during receptor activation.
Collapse
Affiliation(s)
- Michel G Tremblay
- From the Laboratory of Growth and Development, St-Patrick Research Group in Basic Oncology, Cancer Division of the Québec University Hospital Research Centre, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Edifice St Patrick, 9 rue McMahon, Québec, Québec G1R 3S3, Canada
| | - Chelsea Herdman
- From the Laboratory of Growth and Development, St-Patrick Research Group in Basic Oncology, Cancer Division of the Québec University Hospital Research Centre, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Edifice St Patrick, 9 rue McMahon, Québec, Québec G1R 3S3, Canada
| | - François Guillou
- From the Laboratory of Growth and Development, St-Patrick Research Group in Basic Oncology, Cancer Division of the Québec University Hospital Research Centre, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Edifice St Patrick, 9 rue McMahon, Québec, Québec G1R 3S3, Canada
| | - Prakash K Mishra
- From the Laboratory of Growth and Development, St-Patrick Research Group in Basic Oncology, Cancer Division of the Québec University Hospital Research Centre, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Edifice St Patrick, 9 rue McMahon, Québec, Québec G1R 3S3, Canada
| | - Joëlle Baril
- From the Laboratory of Growth and Development, St-Patrick Research Group in Basic Oncology, Cancer Division of the Québec University Hospital Research Centre, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Edifice St Patrick, 9 rue McMahon, Québec, Québec G1R 3S3, Canada
| | - Sabrina Bellenfant
- From the Laboratory of Growth and Development, St-Patrick Research Group in Basic Oncology, Cancer Division of the Québec University Hospital Research Centre, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Edifice St Patrick, 9 rue McMahon, Québec, Québec G1R 3S3, Canada
| | - Tom Moss
- From the Laboratory of Growth and Development, St-Patrick Research Group in Basic Oncology, Cancer Division of the Québec University Hospital Research Centre, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Edifice St Patrick, 9 rue McMahon, Québec, Québec G1R 3S3, Canada
| |
Collapse
|
79
|
Di Giovanni V, Walker KA, Bushnell D, Schaefer C, Sims-Lucas S, Puri P, Bates CM. Fibroblast growth factor receptor-Frs2α signaling is critical for nephron progenitors. Dev Biol 2015; 400:82-93. [PMID: 25641696 DOI: 10.1016/j.ydbio.2015.01.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/13/2015] [Accepted: 01/19/2015] [Indexed: 02/07/2023]
Abstract
Previous studies using transgenic Pax3cre mice have revealed roles for fibroblast growth factor receptors (Fgfrs) and Fgfr substrate 2α (Frs2α) signaling in early metanephric mesenchyme patterning and in ureteric morphogenesis. The role of Fgfr/Frs2α signaling in nephron progenitors is unknown. Thus, we generated mouse models using BAC transgenic Six2EGFPcre (Six2cre) mediated deletion of Fgfrs and/or Frs2α in nephron progenitors. Six2cre mediated deletion of Fgfr1 or Fgfr2 alone led to no obvious kidney defects. Six2creFgfr1(flox/flox)Fgfr2(flox/flox) (Fgfr1/2(NP-/-)) mice generate a discernable kidney; however, they develop nephron progenitor depletion starting at embryonic day 12.5 (E12.5) and later demonstrate severe cystic dysplasia. To determine the role of Frs2α signaling downstream of Fgfr2 in Fgfr1/2(NP-/-) mice, we generated Six2cre(,)Fgfr1(flox/flox)Fgfr2(LR/LR) (Fgfr1(NP-/-)Fgfr2(LR/LR)) mice that have point mutations in the Frs2α binding site of Fgfr2. Like Fgfr1/2(NP-/-) mice, Fgfr1(NP-/-)Fgfr2(LR/LR) develop nephron progenitor depletion, but it does not start until E14.5 and older mice have less severe cystic dysplasia than Fgfr1/2(NP-/-) To determine the role of Frs2α alone in nephron progenitors, we generated Six2creFrs2'A(flox/flox) (Frs2a(NP-/-)) mice. Frs2a(NP-/-)mice also develop nephron progenitor depletion and renal cysts, although these occurred later and were less severe than in the other Six2cre mutant mice. The nephron progenitor loss in all Six2cre mutant lines was associated with decreased Cited1 expression and increased apoptosis versus controls. FAC-sorted nephron progenitors in Six2cre Frs2'A(flox/flox) mice demonstrated evidence of increased Notch activity versus controls, which likely drives the progenitor defects. Thus, Fgfr1 and Fgfr2 have synergistic roles in maintaining nephron progenitors; furthermore, Fgfr signaling in nephron progenitors appears to be mediated predominantly by Frs2α.
Collapse
Affiliation(s)
- Valeria Di Giovanni
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Kenneth A Walker
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Daniel Bushnell
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Caitlin Schaefer
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Sunder Sims-Lucas
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Pawan Puri
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Carlton M Bates
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA; Rangos Research Center, Children׳s Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, USA.
| |
Collapse
|
80
|
Ding X, Yang Z, Zhou F, Hu X, Zhou C, Luo C, He Z, Liu Q, Li H, Yan F, Wang F, Xiang S, Zhang J. Human intersectin 2 (ITSN2) binds to Eps8 protein and enhances its degradation. BMB Rep 2014; 45:183-8. [PMID: 22449706 DOI: 10.5483/bmbrep.2012.45.3.183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Participates in actin remodeling through Rac and receptor endocytosis via Rab5. Here, we used yeast two-hybrid system with Eps8 as bait to screen a human brain cDNA library. ITSN2 was identified as the novel binding factor of Eps8. The interaction between ITSN2 and Eps8 was demonstrated by the in vivo co-immunoprecipitation and colocalization assays and the in vitro GST pull-down assays. Furthermore, we mapped the interaction domains to the region between amino acids 260-306 of Eps8 and the coiled-coil domain of ITSN2. In addition, protein stability assays and immunofluorescence analysis showed ITSN2 overexpression induced the degradation of Eps8 proteins, which was markedly alleviated with the lysosome inhibitor NH4Cl treatment. Taken together, our results suggested ITSN2 interacts with Eps8 and stimulates the degradation of Eps8 proteins.
Collapse
Affiliation(s)
- Xiaofeng Ding
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Kim YJ, Bahn M, Kim YH, Shin JY, Cheong SW, Ju BG, Kim WS, Yeo CY. Xenopus laevis FGF receptor substrate 3 (XFrs3) is important for eye development and mediates Pax6 expression in lens placode through its Shp2-binding sites. Dev Biol 2014; 397:129-39. [PMID: 25446028 DOI: 10.1016/j.ydbio.2014.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 10/13/2014] [Accepted: 10/26/2014] [Indexed: 10/24/2022]
Abstract
Members of the fibroblast growth factor (FGF) family play important roles during various developmental processes including eye development. FRS (FGF receptor substrate) proteins bind to FGFR and serve as adapters for coordinated assembly of multi-protein complexes involved in Ras/MAPK and PI3 kinase/Akt pathways. Here, we identified Xenopus laevis Frs3 (XFrs3), a homolog of vertebrate Frs3, and investigated its roles during embryogenesis. XFrs3 is expressed maternally and zygotically with specific expression patterns throughout the early development. Knockdown of XFrs3 using a specific antisense morpholino oligonucleotide (MO) caused reduction of Pax6 expression in the lens placode, and defects in the eye ranging from microphthalmia to anophthalmia. XFrs3 MO-induced defects were alleviated by wild type XFrs3 or a mutant XFrs3 (XFrs3-4YF), in which the putative tyrosine phosphorylation sites served as Grb2-binding sites are mutated. However, another XFrs3 mutant (XFrs3-2YF), in which the putative Shp2-binding sites are mutated, could not rescue the defects of XFrs3 morphants. In addition, we found that XFrs3 is important for FGF or IGF-induced ERK activation in ectodermal tissue. Taken together, our results suggest that signaling through Shp2-binding sites of XFrs3 is necessary for the eye development in Xenopus laevis.
Collapse
Affiliation(s)
- Yeon-Jin Kim
- Department of Life Science and Global Top5 Research Program, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Minjin Bahn
- Department of Life Science and Global Top5 Research Program, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Yong Hwan Kim
- Department of Life Sciences, Sogang University, Seoul 121-742, Republic of Korea
| | - Jee-Yoon Shin
- Department of Life Sciences, Sogang University, Seoul 121-742, Republic of Korea
| | - Seon-Woo Cheong
- Department of Biology, Changwon National University, Changwon 614-773, Republic of Korea
| | - Bong-Gun Ju
- Department of Life Sciences, Sogang University, Seoul 121-742, Republic of Korea
| | - Won-Sun Kim
- Department of Life Sciences, Sogang University, Seoul 121-742, Republic of Korea.
| | - Chang-Yeol Yeo
- Department of Life Science and Global Top5 Research Program, Ewha Womans University, Seoul 120-750, Republic of Korea.
| |
Collapse
|
82
|
Mazzola CR, Siddiqui KM, Billia M, Chin J. Dovitinib: rationale, preclinical and early clinical data in urothelial carcinoma of the bladder. Expert Opin Investig Drugs 2014; 23:1553-62. [PMID: 25284004 DOI: 10.1517/13543784.2014.966900] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Bladder cancer (BC) is the third and fifth cancer in men in terms of incidence and mortality in the US. Overexpression and mutations of fibroblast growth factor receptor 3 (FGFR3) are frequently found in BC and can represent a very interesting therapeutic target. Different FGFR3-targeted strategies have been investigated through in vitro and in vivo settings, including FGFR3 tyrosine kinase inhibitors such as dovitinib . AREAS COVERED The authors review the data that provide a scientific rationale for FGFR3-targeted therapy in BC. They also provide an evaluation of the currently available in vitro and in vivo data on the use of dovitinib in BC patients. EXPERT OPINION The development and progression of BC rely on a very complex signaling network that involves many different receptors aside from FGFR3 and VEGFR2. The involved signaling network can also be very different from one BC to the other, and can also evolve through time in the same patient. Inhibiting only one single target may thus not be sufficient to achieve a complete downstream oncogenic signaling blockage. Additionally, in vitro data on the use of neutralizing monoclonal antibodies targeting FGFR3 show that it can be a more efficient strategy to reach the same goal, with the potential advantage of less toxicity.
Collapse
Affiliation(s)
- Clarisse R Mazzola
- Western University, Division of Urology and Division of Surgical Oncology , London, Ontario , Canada +519 685 8451 ; +519 685 8455 ;
| | | | | | | |
Collapse
|
83
|
Zhou L, Talebian A, Meakin SO. The signaling adapter, FRS2, facilitates neuronal branching in primary cortical neurons via both Grb2- and Shp2-dependent mechanisms. J Mol Neurosci 2014; 55:663-77. [PMID: 25159185 DOI: 10.1007/s12031-014-0406-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 08/15/2014] [Indexed: 12/15/2022]
Abstract
The neurotrophins are a family of closely related growth factors that regulate proliferation and differentiation in the developing and mature nervous systems. Neurotrophins stimulate a family of receptor tyrosine kinases (Trk receptors) and utilize an intracellular docking protein termed fibroblast growth factor (FGF) receptor substrate 2 (FRS2) as a major downstream adapter to activate Ras, phosphatidylinositide 3-kinase (PI3K), and mitogen-activated protein kinase (MAPK) signaling cascades. The goals of this study were twofold: first, to investigate the complexity of neurotrophin-induced FRS2 interactions in primary cortical neurons and to determine which pathway(s) are important in regulating neuronal growth and, second, to determine whether the related signaling adapter, FRS3, stimulates neuron growth comparable to FRS2. We find that neurotrophin treatment of primary cortical neurons stimulates the tyrosine phosphorylation of FRS2 and the subsequent recruitment of Shp2, Grb2, and Gab2. With FRS2 mutants deficient in Grb2 or Shp2 binding, we demonstrate that FRS2 binds Gab1 and Gab2 through Grb2, providing an alternative route to activate PI3 kinase and Shp2. Using recombinant adenoviruses expressing FRS2, we demonstrate that FRS2 overexpression promotes neurite outgrowth and branching in cortical neurons relative to controls. In contrast, overexpression of FRS3 does not stimulate neuronal growth. Moreover, we find that while loss of Shp2, but not Grb2, reduces brain-derived neurotrophic factor (BDNF)-induced MAPK activation, the loss of either pathway impairs neuronal growth. Collectively, these experiments demonstrate that FRS2 functions as an adapter of a multiprotein complex that is activated by the Trk receptors and that the activation of both Grb2- and Shp2-dependent pathways facilitates cortical neuronal growth.
Collapse
Affiliation(s)
- Li Zhou
- Laboratory of Neural Signaling, Molecular Medicine Research Group, The Robarts Research Institute, 1151 Richmond St. N, London, Ontario, N6A 5B7, Canada
| | | | | |
Collapse
|
84
|
Lu H, Han YJ, Xu JD, Xing WM, Chen J. Proteomic characterization of acyclovir-induced nephrotoxicity in a mouse model. PLoS One 2014; 9:e103185. [PMID: 25055032 PMCID: PMC4108384 DOI: 10.1371/journal.pone.0103185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 06/30/2014] [Indexed: 02/07/2023] Open
Abstract
Acyclovir (ACV) is an effective and widely used antiviral agent. However, its clinical application is limited by severe nephrotoxicity. We assessed ACV-induced nephrotoxicity and identified the differentially expressed proteins using mass spectrometry-based proteomic analysis. In total, 30 ICR mice were intraperitoneally administrated ACV (150 or 600 mg/kg per day) for 9 days. After administration of ACV, levels of serum creatinine and urea nitrogen increased significantly. In addition, mouse kidneys exhibited histopathological changes and reduced expression levels of vascular endothelial growth factor (VEGF) and its receptor VEGFR2. In the proteomic analysis, more than 1,000 proteins were separated by two-dimensional polyacrylamide gel electrophoresis, and a total of 20 proteins were up- or down-regulated in the ACV group compared with the saline group. Among these, six proteins (MHC class II antigen, glyoxalase 1, peroxiredoxin 1, αB-crystallin, fibroblast growth factor receptor 1-IIIb, and cytochrome c oxidase subunit Vb) were identified in association with ACV-induced nephrotoxicity. These findings were confirmed by Western blotting analysis. The differential expression levels of α-BC, Prx1, Glo I and CcO Vb suggest that oxidative damage and mitochondrial injury may be involved in ACV-induced nephrotoxicity. Furthermore, VEGF and FGF may play a role in tissue repair and the restoration process following ACV nephrotoxicity.
Collapse
Affiliation(s)
- Hong Lu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Ya-Juan Han
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jia-Dong Xu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Wen-Min Xing
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jie Chen
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| |
Collapse
|
85
|
Grusch M, Schelch K, Riedler R, Reichhart E, Differ C, Berger W, Inglés-Prieto Á, Janovjak H. Spatio-temporally precise activation of engineered receptor tyrosine kinases by light. EMBO J 2014; 33:1713-26. [PMID: 24986882 DOI: 10.15252/embj.201387695] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are a large family of cell surface receptors that sense growth factors and hormones and regulate a variety of cell behaviours in health and disease. Contactless activation of RTKs with spatial and temporal precision is currently not feasible. Here, we generated RTKs that are insensitive to endogenous ligands but can be selectively activated by low-intensity blue light. We screened light-oxygen-voltage (LOV)-sensing domains for their ability to activate RTKs by light-activated dimerization. Incorporation of LOV domains found in aureochrome photoreceptors of stramenopiles resulted in robust activation of the fibroblast growth factor receptor 1 (FGFR1), epidermal growth factor receptor (EGFR) and rearranged during transfection (RET). In human cancer and endothelial cells, light induced cellular signalling with spatial and temporal precision. Furthermore, light faithfully mimicked complex mitogenic and morphogenic cell behaviour induced by growth factors. RTKs under optical control (Opto-RTKs) provide a powerful optogenetic approach to actuate cellular signals and manipulate cell behaviour.
Collapse
Affiliation(s)
- Michael Grusch
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Karin Schelch
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Robert Riedler
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Eva Reichhart
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Christopher Differ
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Álvaro Inglés-Prieto
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Harald Janovjak
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| |
Collapse
|
86
|
Uren RT, Turnley AM. Regulation of neurotrophin receptor (Trk) signaling: suppressor of cytokine signaling 2 (SOCS2) is a new player. Front Mol Neurosci 2014; 7:39. [PMID: 24860421 PMCID: PMC4030161 DOI: 10.3389/fnmol.2014.00039] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/21/2014] [Indexed: 12/17/2022] Open
Abstract
The classic neurotrophins Nerve Growth Factor (NGF), Brain Derived Neurotrophic Factor (BDNF) and Neurotrophins NT-3 and NT-4 are well known to regulate various aspects of neuronal differentiation, survival and growth. They do this by binding to their cognate receptors, members of the Tropomyosin-related kinase (Trk) receptor tyrosine kinase family, namely TrkA, TrkB, and TrkC. These receptors are then internalized and localized to different cellular compartments, where signal transduction occurs. Conversely, members of the suppressor of cytokine signaling (SOCS) family are best known as negative regulators of signaling via the JAK/STAT pathway. Some members of the family, and in particular SOCS2, have roles in the nervous system that at least partially overlap with that of neurotrophins, namely neuronal differentiation and neurite outgrowth. Recent evidence suggests that SOCS2 is a novel regulator of NGF signaling, altering TrkA cellular localization and downstream signaling to affect neurite growth but not neuronal survival. This review first discusses regulation of Trk receptor signaling, followed by the role of SOCS2 in the nervous system and finishes with a discussion of possible mechanisms by which SOCS2 may regulate TrkA function.
Collapse
Affiliation(s)
- Rachel T Uren
- Neural Regeneration Laboratory, Centre for Neuroscience Research and Department of Anatomy and Neuroscience, The University of Melbourne Melbourne, VIC, Australia
| | - Ann M Turnley
- Neural Regeneration Laboratory, Centre for Neuroscience Research and Department of Anatomy and Neuroscience, The University of Melbourne Melbourne, VIC, Australia
| |
Collapse
|
87
|
The docking protein FRS2α is a critical regulator of VEGF receptors signaling. Proc Natl Acad Sci U S A 2014; 111:5514-9. [PMID: 24706887 DOI: 10.1073/pnas.1404545111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) signal via their cognate receptor tyrosine kinases designated VEGFR1-3. We report that the docking protein fibroblast growth factor receptor substrate 2 (FRS2α) plays a critical role in cell signaling via these receptors. In vitro FRS2α regulates VEGF-A and VEGF-C-dependent activation of extracellular signal-regulated receptor kinase signaling and blood and lymphatic endothelial cells migration and proliferation. In vivo endothelial-specific deletion of FRS2α results in the profound impairment of postnatal vascular development and adult angiogenesis, lymphangiogenesis, and arteriogenesis. We conclude that FRS2α is a previously unidentified component of VEGF receptors signaling.
Collapse
|
88
|
Youssef G, Gillett C, Agbaje O, Crompton T, Montano X. Phosphorylation of NTRK1 at Y674/Y675 induced by TP53-dependent repression of PTPN6 expression: a potential novel prognostic marker for breast cancer. Mod Pathol 2014; 27:361-74. [PMID: 23948750 DOI: 10.1038/modpathol.2013.129] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/29/2013] [Accepted: 05/30/2013] [Indexed: 12/29/2022]
Abstract
We have identified a ligand-independent mechanism whereby the tumor suppressor, TP53, induces nerve growth factor receptor, NTRK1, phosphorylation at Y674/Y675 (NTRK1-pY674/pY675), via the repression of the NTRK1-phosphatase, PTPN6. This results in suppression of breast cancer cell proliferation. In this investigation, we aimed to establish whether perturbation of the wild-type TP53-NTRK1-pY674/pY675-PTPN6 pathway has an impact on disease-free survival of breast cancer patients without neo-adjuvant treatment. A total of 308 tumor samples were stained for NTRK1, NTRK1-pY674/pY675, PTPN6, and TP53 expression. Association between expression levels and disease-free survival was determined by the univariate/multivariate and Kaplan-Meir methods of analysis. DNA from tumors was sequenced to identify mutant or wild-type TP53. Tumors expressing NTRK1-pY674/pY675 but with undetectable or low levels of PTPN6 and TP53 were associated with prolonged 5, 10, and 15 years' disease-free survival by 48%, 36%, and 37%, respectively, in the multivariate analysis (P<0.05). A similar result was observed in tumors expressing wild-type TP53, NTRK1-pY674/pY675, and low or undetectable levels of PTPN6. Given that estrogen receptor-positive breast cancers encode wild-type TP53, we analyzed this expression pattern in these tumors. Multivariate analysis showed that it was significantly and independently predictive of prolonged survival by 66%, 70%, and 84%, respectively, (P<0.05). The Kaplan-Meir method demonstrated that NTRK1-pY674/pY675 together with undetectable or low levels of PTPN6 correlated with 59% probability of disease-free survival (median survival 15 years), compared with 7% probability of disease-free survival (median survival 4.5 years) when absent. In luminal A tumors, the presence of this pattern was estimated to have a 61% probability of disease-free survival (median survival 15 years), compared with 6% probability of disease-free survival (median survival 3 years) when it was absent. These results strongly suggest that expression of NTRK1-pY674/pY675 together with wild-type TP53 and low levels of PTPN6 expression are predictors of improved disease-free survival and that they could be useful biomarkers to predict clinical outcome.
Collapse
Affiliation(s)
- Gehad Youssef
- 1] Immunobiology Unit, UCL, Institute of Child Health, London, UK [2] Molecular Hematology and Cancer Biology Unit, UCL, Institute of Child Health, London, UK
| | - Cheryl Gillett
- Breast Tissue & Data Bank, Division of Cancer Studies, King's College London, Guys Hospital, London, UK
| | - Orunsola Agbaje
- Cancer Epidemiology Group, Division of Cancer Studies, King's College London, Guys Hospital, London, UK
| | - Tessa Crompton
- Immunobiology Unit, UCL, Institute of Child Health, London, UK
| | - Ximena Montano
- 1] Immunobiology Unit, UCL, Institute of Child Health, London, UK [2] Molecular Hematology and Cancer Biology Unit, UCL, Institute of Child Health, London, UK [3] School of Health and Social Work, Department of Allied Health Professions and Midwifery University of Hertfordshire, Hatfield, UK
| |
Collapse
|
89
|
Flotillins in receptor tyrosine kinase signaling and cancer. Cells 2014; 3:129-49. [PMID: 24709906 PMCID: PMC3980747 DOI: 10.3390/cells3010129] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 02/11/2014] [Accepted: 02/12/2014] [Indexed: 01/23/2023] Open
Abstract
Flotillins are highly conserved proteins that localize into specific cholesterol rich microdomains in cellular membranes. They have been shown to be associated with, for example, various signaling pathways, cell adhesion, membrane trafficking and axonal growth. Recent findings have revealed that flotillins are frequently overexpressed in various types of human cancers. We here review the suggested functions of flotillins during receptor tyrosine kinase signaling and in cancer. Although flotillins have been implicated as putative cancer therapy targets, we here show that great caution is required since flotillin ablation may result in effects that increase instead of decrease the activity of specific signaling pathways. On the other hand, as flotillin overexpression appears to be related with metastasis formation in certain cancers, we also discuss the implications of these findings for future therapy aspects.
Collapse
|
90
|
Fibroblast growth factor receptor 4 (FGFR4): a targetable regulator of drug resistance in colorectal cancer. Cell Death Dis 2014; 5:e1046. [PMID: 24503538 PMCID: PMC3944229 DOI: 10.1038/cddis.2014.10] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/21/2013] [Accepted: 12/23/2013] [Indexed: 12/18/2022]
Abstract
The discovery of underlying mechanisms of drug resistance, and the development of novel agents to target these pathways, is a priority for patients with advanced colorectal cancer (CRC). We previously undertook a systems biology approach to design a functional genomic screen and identified fibroblast growth factor receptor 4 (FGFR4) as a potential mediator of drug resistance. The aim of this study was to examine the role of FGFR4 in drug resistance using RNAi and the small-molecule inhibitor BGJ398 (Novartis). We found that FGFR4 is highly expressed at the RNA and protein levels in colon cancer tumour tissue compared with normal colonic mucosa and other tumours. Silencing of FGFR4 reduced cell viability in a panel of colon cancer cell lines and increased caspase-dependent apoptosis. A synergistic interaction was also observed between FGFR4 silencing and 5-fluorouracil (5-FU) and oxaliplatin chemotherapy in colon cancer cell lines. Mechanistically, FGFR4 silencing decreased activity of the pro-survival STAT3 transcription factor and expression of the anti-apoptotic protein c-FLIP. Furthermore, silencing of STAT3 resulted in downregulation of c-FLIP protein expression, suggesting that FGFR4 may regulate c-FLIP expression via STAT3. A similar phenotype and downstream pathway changes were observed following FGFR4 silencing in cell lines resistant to 5-FU, oxaliplatin and SN38 and upon exposure of parental cells to the FGFR small-molecule inhibitor BGJ398. Our results indicate that FGFR4 is a targetable regulator of chemo-resistance in CRC, and hence inhibiting FGFR4 in combination with 5-FU and oxaliplatin is a potential therapeutic strategy for this disease.
Collapse
|
91
|
Salazar L, Kashiwada T, Krejci P, Meyer AN, Casale M, Hallowell M, Wilcox WR, Donoghue DJ, Thompson LM. Fibroblast growth factor receptor 3 interacts with and activates TGFβ-activated kinase 1 tyrosine phosphorylation and NFκB signaling in multiple myeloma and bladder cancer. PLoS One 2014; 9:e86470. [PMID: 24466111 PMCID: PMC3900522 DOI: 10.1371/journal.pone.0086470] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 12/09/2013] [Indexed: 12/31/2022] Open
Abstract
Cancer is a major public health problem worldwide. In the United States alone, 1 in 4 deaths is due to cancer and for 2013 a total of 1,660,290 new cancer cases and 580,350 cancer-related deaths are projected. Comprehensive profiling of multiple cancer genomes has revealed a highly complex genetic landscape in which a large number of altered genes, varying from tumor to tumor, impact core biological pathways and processes. This has implications for therapeutic targeting of signaling networks in the development of treatments for specific cancers. The NFκB transcription factor is constitutively active in a number of hematologic and solid tumors, and many signaling pathways implicated in cancer are likely connected to NFκB activation. A critical mediator of NFκB activity is TGFβ-activated kinase 1 (TAK1). Here, we identify TAK1 as a novel interacting protein and target of fibroblast growth factor receptor 3 (FGFR3) tyrosine kinase activity. We further demonstrate that activating mutations in FGFR3 associated with both multiple myeloma and bladder cancer can modulate expression of genes that regulate NFκB signaling, and promote both NFκB transcriptional activity and cell adhesion in a manner dependent on TAK1 expression in both cancer cell types. Our findings suggest TAK1 as a potential therapeutic target for FGFR3-associated cancers, and other malignancies in which TAK1 contributes to constitutive NFκB activation.
Collapse
MESH Headings
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Adhesion
- Cell Proliferation
- Gene Expression Profiling
- Humans
- Immunoprecipitation
- MAP Kinase Kinase Kinases/genetics
- MAP Kinase Kinase Kinases/metabolism
- Multiple Myeloma/genetics
- Multiple Myeloma/metabolism
- Multiple Myeloma/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Oligonucleotide Array Sequence Analysis
- Peptide Fragments
- Phosphorylation
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Tumor Cells, Cultured
- Two-Hybrid System Techniques
- Tyrosine/metabolism
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/pathology
Collapse
Affiliation(s)
- Lisa Salazar
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California, United States of America
| | - Tamara Kashiwada
- Department of Biological Chemistry, University of California Irvine, Irvine, California, United States of America
| | - Pavel Krejci
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Institute of Experimental Biology, Masaryk University and Department of Cytokinetics, Institute of Biophysics AS CR, v.v.i., Brno, Czech Republic
- Department of Pediatrics, UCLA School of Medicine, Los Angeles, California, United States of America
| | - April N. Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Malcolm Casale
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
| | - Matthew Hallowell
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California, United States of America
| | - William R. Wilcox
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Pediatrics, UCLA School of Medicine, Los Angeles, California, United States of America
| | - Daniel J. Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
- Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Leslie Michels Thompson
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California, United States of America
- Department of Biological Chemistry, University of California Irvine, Irvine, California, United States of America
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
92
|
Li H, Tao C, Cai Z, Hertzler-Schaefer K, Collins TN, Wang F, Feng GS, Gotoh N, Zhang X. Frs2α and Shp2 signal independently of Gab to mediate FGF signaling in lens development. J Cell Sci 2013; 127:571-82. [PMID: 24284065 DOI: 10.1242/jcs.134478] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Fibroblast growth factor (FGF) signaling requires a plethora of adaptor proteins to elicit downstream responses, but the functional significances of these docking proteins remain controversial. In this study, we used lens development as a model to investigate Frs2α and its structurally related scaffolding proteins, Gab1 and Gab2, in FGF signaling. We show that genetic ablation of Frs2α alone has a modest effect, but additional deletion of tyrosine phosphatase Shp2 causes a complete arrest of lens vesicle development. Biochemical evidence suggests that this Frs2α-Shp2 synergy reflects their epistatic relationship in the FGF signaling cascade, as opposed to compensatory or parallel functions of these two proteins. Genetic interaction experiments further demonstrate that direct binding of Shp2 to Frs2α is necessary for activation of ERK signaling, whereas constitutive activation of either Shp2 or Kras signaling can compensate for the absence of Frs2α in lens development. By contrast, knockout of Gab1 and Gab2 failed to disrupt FGF signaling in vitro and lens development in vivo. These results establish the Frs2α-Shp2 complex as the key mediator of FGF signaling in lens development.
Collapse
Affiliation(s)
- Hongge Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Huang J, Feng L, An H, Zhang T. Construction of Y376C-FGFR4 eukaryotic expression plasmid and its biological activity in HEK293 cell. Acta Biochim Biophys Sin (Shanghai) 2013; 45:889-92. [PMID: 23985304 DOI: 10.1093/abbs/gmt091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Jing Huang
- School of Pharmaceutical Sciences, Xi'an Jiaotong University, Xi'an 710061, China
| | | | | | | |
Collapse
|
94
|
Kelleher FC, O'Sullivan H, Smyth E, McDermott R, Viterbo A. Fibroblast growth factor receptors, developmental corruption and malignant disease. Carcinogenesis 2013; 34:2198-205. [PMID: 23880303 DOI: 10.1093/carcin/bgt254] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Fibroblast growth factors (FGF) are a family of ligands that bind to four different types of cell surface receptor entitled, FGFR1, FGFR2, FGFR3 and FGFR4. These receptors differ in their ligand binding affinity and tissue distribution. The prototypical receptor structure is that of an extracellular region comprising three immunoglobulin (Ig)-like domains, a hydrophobic transmembrane segment and a split intracellular tyrosine kinase domain. Alternative gene splicing affecting the extracellular third Ig loop also creates different receptor isoforms entitled FGFRIIIb and FGFRIIIc. Somatic fibroblast growth factor receptor (FGFR) mutations are implicated in different types of cancer and germline FGFR mutations occur in developmental syndromes particularly those in which craniosynostosis is a feature. The mutations found in both conditions are often identical. Many somatic FGFR mutations in cancer are gain-of-function mutations of established preclinical oncogenic potential. Gene amplification can also occur with 19-22% of squamous cell lung cancers for example having amplification of FGFR1. Ontologic comparators can be informative such as aberrant spermatogenesis being implicated in both spermatocytic seminomas and Apert syndrome. The former arises from somatic FGFR3 mutations and Apert syndrome arises from germline FGFR2 mutations. Finally, therapeutics directed at inhibiting the FGF/FGFR interaction are a promising subject for clinical trials.
Collapse
Affiliation(s)
- Fergal C Kelleher
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria VIC8006, Australia
| | | | | | | | | |
Collapse
|
95
|
Posttranslational protein knockdown coupled to receptor tyrosine kinase activation with phosphoPROTACs. Proc Natl Acad Sci U S A 2013; 110:8942-7. [PMID: 23674677 DOI: 10.1073/pnas.1217206110] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Posttranslational knockdown of a specific protein is an attractive approach for examining its function within a system. Here we introduce phospho-dependent proteolysis targeting chimeras (phosphoPROTACs), a method to couple the conditional degradation of targeted proteins to the activation state of particular kinase-signaling pathways. We generated two phosphoPROTACs that couple the tyrosine phosphorylation sequences of either the nerve growth factor receptor, TrkA (tropomyosin receptor kinase A), or the neuregulin receptor, ErbB3 (erythroblastosis oncogene B3), with a peptide ligand for the E3 ubiquitin ligase von Hippel Lindau protein. These phosphoPROTACs recruit either the neurotrophic signaling effector fibroblast growth factor receptor substrate 2α or the survival-promoting phosphatidylinositol-3-kinase, respectively, to be ubiquitinated and degraded upon activation of specific receptor tyrosine kinases and phosphorylation of the phosphoPROTACs. We demonstrate the ability of these phosphoPROTACs to suppress the short- and long-term effects of their respective activating receptor tyrosine kinase pathways both in vitro and in vivo. In addition, we show that activation of phosphoPROTACs is entirely dependent on their kinase-mediated phosphorylation, as phenylalanine-containing null variants are inactive. Furthermore, stimulation of unrelated growth factor receptors does not induce target protein knockdown. Although comparable in efficiency to RNAi, this approach has the added advantage of providing a degree of temporal and dosing control as well as cell-type selectivity unavailable using nucleic acid-based strategies. By varying the autophosphorylation sequence of a phosphoPROTAC, it is conceivable that other receptor tyrosine kinase/effector pairings could be similarly exploited to achieve other biological effects.
Collapse
|
96
|
The antiaging protein Klotho enhances oligodendrocyte maturation and myelination of the CNS. J Neurosci 2013; 33:1927-39. [PMID: 23365232 DOI: 10.1523/jneurosci.2080-12.2013] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We have previously shown that myelin abnormalities characterize the normal aging process of the brain and that an age-associated reduction in Klotho is conserved across species. Predominantly generated in brain and kidney, Klotho overexpression extends life span, whereas loss of Klotho accelerates the development of aging-like phenotypes. Although the function of Klotho in brain is unknown, loss of Klotho expression leads to cognitive deficits. We found significant effects of Klotho on oligodendrocyte functions, including induced maturation of rat primary oligodendrocytic progenitor cells (OPCs) in vitro and myelination. Phosphoprotein analysis indicated that Klotho's downstream effects involve Akt and ERK signal pathways. Klotho increased OPC maturation, and inhibition of Akt or ERK function blocked this effect on OPCs. In vivo studies of Klotho knock-out mice and control littermates revealed that knock-out mice have a significant reduction in major myelin protein and gene expression. By immunohistochemistry, the number of total and mature oligodendrocytes was significantly lower in Klotho knock-out mice. Strikingly, at the ultrastructural level, Klotho knock-out mice exhibited significantly impaired myelination of the optic nerve and corpus callosum. These mice also displayed severe abnormalities at the nodes of Ranvier. To decipher the mechanisms by which Klotho affects oligodendrocytes, we used luciferase pathway reporters to identify the transcription factors involved. Together, these studies provide novel evidence for Klotho as a key player in myelin biology, which may thus be a useful therapeutic target in efforts to protect brain myelin against age-dependent changes and promote repair in multiple sclerosis.
Collapse
|
97
|
Meister M, Tomasovic A, Banning A, Tikkanen R. Mitogen-Activated Protein (MAP) Kinase Scaffolding Proteins: A Recount. Int J Mol Sci 2013; 14:4854-84. [PMID: 23455463 PMCID: PMC3634400 DOI: 10.3390/ijms14034854] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/17/2013] [Accepted: 02/21/2013] [Indexed: 12/20/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK) pathway is the canonical signaling pathway for many receptor tyrosine kinases, such as the Epidermal Growth Factor Receptor. Downstream of the receptors, this pathway involves the activation of a kinase cascade that culminates in a transcriptional response and affects processes, such as cell migration and adhesion. In addition, the strength and duration of the upstream signal also influence the mode of the cellular response that is switched on. Thus, the same components can in principle coordinate opposite responses, such as proliferation and differentiation. In recent years, it has become evident that MAPK signaling is regulated and fine-tuned by proteins that can bind to several MAPK signaling proteins simultaneously and, thereby, affect their function. These so-called MAPK scaffolding proteins are, thus, important coordinators of the signaling response in cells. In this review, we summarize the recent advances in the research on MAPK/extracellular signal-regulated kinase (ERK) pathway scaffolders. We will not only review the well-known members of the family, such as kinase suppressor of Ras (KSR), but also put a special focus on the function of the recently identified or less studied scaffolders, such as fibroblast growth factor receptor substrate 2, flotillin-1 and mitogen-activated protein kinase organizer 1.
Collapse
Affiliation(s)
- Melanie Meister
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany; E-Mails: (M.M.); (A.B.)
| | - Ana Tomasovic
- Department of Molecular Hematology, University of Frankfurt, Medical School, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; E-Mail:
| | - Antje Banning
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany; E-Mails: (M.M.); (A.B.)
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany; E-Mails: (M.M.); (A.B.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-641-9947-420; Fax: +49-641-9947-429
| |
Collapse
|
98
|
Irschick R, Trost T, Karp G, Hausott B, Auer M, Claus P, Klimaschewski L. Sorting of the FGF receptor 1 in a human glioma cell line. Histochem Cell Biol 2013; 139:135-48. [PMID: 22903848 DOI: 10.1007/s00418-012-1009-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2012] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factor receptor 1 (FGFR1) is a receptor tyrosine kinase promoting tumor growth in a variety of cancers, including glioblastoma. Binding of FGFs triggers the intracellular Ras/Raf/ERK signaling pathway leading to cell proliferation. Down-regulation of FGFR1 and, consequently, inactivation of its signaling pathways represent novel treatment strategies for glioblastoma. In this study, we investigated the internalization and endocytic trafficking of FGFR1 in the human glioma cell line U373. Stimulation with FGF-2 induced cell rounding accompanied by increased BrdU and pERK labeling. The overexpression of FGFR1 (without FGF treatment) resulted in enhanced phosphorylated FGFR1 suggesting receptor autoactivation. Labeled ligand (FGF-2-Cy5.5) was endocytosed in a clathrin- and caveolin-dependent manner. About 25 % of vesicles carrying fluorescently tagged FGFR1 represented early endosomes, 15 % transferrin-positive recycling endosomes and 40 % Lamp1-positive late endosomal/lysosomal vesicles. Stimulation with FGF-2 increased the colocalization rate in each of these vesicle populations. The treatment with the lysosomal inhibitor leupeptin resulted in FGFR1 accumulation in lysosomes, but did not enhance receptor recycling as observed in neurons. Analysis of vesicle distributions revealed an accumulation of recycling endosomes in the perinuclear region. In conclusion, the shuttling of receptor tyrosine kinases can be directly visualized by overexpression of fluorescently tagged receptors which respond to ligand stimulation and follow the recycling and degradation pathways similarly to their endogenous counterparts.
Collapse
Affiliation(s)
- Regina Irschick
- Division of Neuroanatomy, Medical University Innsbruck, Muellerstrasse 59, 6020 Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
99
|
Madakashira BP, Kobrinski DA, Hancher AD, Arneman EC, Wagner BD, Wang F, Shin H, Lovicu FJ, Reneker LW, Robinson ML. Frs2α enhances fibroblast growth factor-mediated survival and differentiation in lens development. Development 2012; 139:4601-12. [PMID: 23136392 DOI: 10.1242/dev.081737] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Most growth factor receptor tyrosine kinases (RTKs) signal through similar intracellular pathways, but they often have divergent biological effects. Therefore, elucidating the mechanism of channeling the intracellular effect of RTK stimulation to facilitate specific biological responses represents a fundamental biological challenge. Lens epithelial cells express numerous RTKs with the ability to initiate the phosphorylation (activation) of Erk1/2 and PI3-K/Akt signaling. However, only Fgfr stimulation leads to lens fiber cell differentiation in the developing mammalian embryo. Additionally, within the lens, only Fgfrs activate the signal transduction molecule Frs2α. Loss of Frs2α in the lens significantly increases apoptosis and decreases phosphorylation of both Erk1/2 and Akt. Also, Frs2α deficiency decreases the expression of several proteins characteristic of lens fiber cell differentiation, including Prox1, p57(KIP2), aquaporin 0 and β-crystallins. Although not normally expressed in the lens, the RTK TrkC phosphorylates Frs2α in response to binding the ligand NT3. Transgenic lens epithelial cells expressing both TrkC and NT3 exhibit several features characteristic of lens fiber cells. These include elongation, increased Erk1/2 and Akt phosphorylation, and the expression of β-crystallins. All these characteristics of NT3-TrkC transgenic lens epithelial cells depend on Frs2α. Therefore, tyrosine phosphorylation of Frs2α mediates Fgfr-dependent lens cell survival and provides a mechanistic basis for the unique fiber-differentiating capacity of Fgfs on mammalian lens epithelial cells.
Collapse
|
100
|
Bradshaw RA, Chalkley RJ, Biarc J, Burlingame AL. Receptor tyrosine kinase signaling mechanisms: Devolving TrkA responses with phosphoproteomics. Adv Biol Regul 2012; 53:87-96. [PMID: 23266087 DOI: 10.1016/j.jbior.2012.10.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 10/25/2012] [Indexed: 01/07/2023]
Abstract
Receptor tyrosine kinases (RTKs) function through protein kinase entities located in the intracellular domain of each protomer. Following activation by ligand binding, they selectively form phosphotyrosine residues by autocatalytic modification. Some of these sites are involved in maintaining the active conformation of the kinase, while others become docking sites for various adaptor/effector/scaffold proteins, which, after complexing with the receptor, then initiate further responses through cascades of post-translational modifications and the generation of lipid second messengers. Although there is substantial overlap in the pathways and activities stimulated by this superfamily, the molecular features of the endodomains of the sub-families and the moieties that they interact with to perpetrate their signals are surprisingly distinct, which may play a significant role in the regulation and responses of the individual RTK types. Some use large scaffold proteins as the basis for most, if not all, of their signal-generating interactions, while others have numerous receptor endodomain phosphotyrosine sites that are quite overlapping in specificity. The members of the Trk family of receptors each have several tyrosine residues that are phosphorylated following stimulation, including those in the kinase activation loop, but there are only two established sites (Y490 and Y785 on TrkA) that are known to be directly involved in signal propagation. Taking advantage of this limited repertoire of docking sites, we have applied phosphoproteomic methods to dissect the signaling responses of both the native protein and derivatives that have had these two sites modified. Interestingly, a clear subset that was not dependent on either docking site was identified. A comparison with a similar set of data for EGFR indicates a considerable degree of similarity in the downstream signaling profile between these two RTKs.
Collapse
Affiliation(s)
- R A Bradshaw
- Dept. Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
| | | | | | | |
Collapse
|