TGF-Β1-Mediated Transition of Resident Fibroblasts to Cancer-Associated

Total Page:16

File Type:pdf, Size:1020Kb

TGF-Β1-Mediated Transition of Resident Fibroblasts to Cancer-Associated Yoon et al. Oncogenesis (2021) 10:13 https://doi.org/10.1038/s41389-021-00302-5 Oncogenesis ARTICLE Open Access TGF-β1-mediated transition of resident fibroblasts to cancer-associated fibroblasts promotes cancer metastasis in gastrointestinal stromal tumor Hyunho Yoon1,2,Chih-MinTang1,2,SudeepBanerjee1,2,3,AntonioL.Delgado1,2,MayraYebra1,2,JacobDavis1,2 and Jason K. Sicklick 1,2 Abstract Cancer-associated fibroblasts (CAFs) are the most abundant cells in the tumor microenvironment. Crosstalk between tumor cells and CAFs contributes to tumor survival in most epithelial cancers. Recently, utilizing gastrointestinal stromal tumor (GIST) as a model for sarcomas, we identified paracrine networks by which CAFs promote tumor progression and metastasis. However, the mechanisms by which CAFs arise in sarcomas remain unclear. Here, RNA sequencing analysis revealed that transforming growth factor-β1 (TGF-β1) is highly expressed in both tumor cells and CAFs. To determine the functional role of TGF-β1, we treated normal gastric fibroblasts (GFs) with recombinant TGF-β1, which caused the GFs to adopt a more stellate morphology, as well as increased the mRNA expression of CAF- mediated genes (CCL2, RAB3B, and TNC) and genes encoding fibroblast growth factors (FGFs). Moreover, while either GIST or CAF conditioned media enhanced the transition from GFs to CAFs, a TGF-β1-blocking antibody attenuated this effect. Transwell migration assays revealed that the TGF-β1-mediated transition from GFs to CAFs enhanced tumor cell migration. This migratory effect was abrogated by an anti-TGF-β1 antibody, suggesting that TGF-β1 secreted from 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; GIST cells or CAFs is associated with GIST migration via GF-to-CAF transition. In addition, the murine spleen-to-liver metastasis model showed that GF pre-treated with TGF-β1 promoted GIST metastasis. Collectively, these findings reveal unappreciated crosstalk among tumor cells, CAFs, and normal resident fibroblasts in the stroma of sarcomas, which enhances a GF-to-CAF transition associated with tumor migration and metastasis. Introduction sunitinib, regorafenib, and ripretinib are only 7.0%, 4.5%, – A gastrointestinal stromal tumor (GIST), the most and 9.4%, respectively5 7. Due to the high rates of common sarcoma, is most often driven by oncogenic KIT recurrence, alternative therapeutic targets are needed to – or PDGFRA mutations1 3. As a result, anti-KIT/PDGFRA effectively treat GIST. tyrosine kinase inhibitors (TKIs) are commonly used to Cancer-associated fibroblasts (CAFs) are the most treat these tumors. However, 50% of metastatic GISTs abundant cells in the tumor microenvironment (TME). treated with imatinib, the first-line treatment, will develop It is well established that crosstalk between epithelial drug-resistance within 20 months of starting therapy cancers and CAFs can promote tumor progression and – [median progression-free survival (PFS) 20.4 month]4. chemoresistance8 10. Specifically, CAFs can promote a The objective response rates for later line anti-GIST TKIs, favorable biological environment for tumor growth by secreting cytokines, chemokines, and growth factors. Over time, the spatial and temporal interactions of tumor cells Correspondence: Jason K. Sicklick ([email protected]) and stromal cells create a symbiotic relationship in which 1 Department of Surgery, Division of Surgical Oncology, University of California, the tumor receives a growth advantage from the CAFs, San Diego, CA, USA 11 2Moores Cancer Center, University of California, San Diego, CA, USA and vice versa . In addition, our previous study showed Full list of author information is available at the end of the article © The Author(s) 2021 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a linktotheCreativeCommons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/. Oncogenesis Yoon et al. Oncogenesis (2021) 10:13 Page 2 of 12 paracrine fibroblastic support in GIST promotes tumor compared mRNA expression of CAF-mediated genes growth and metastasis12. These studies suggest that CAFs (CCL2, RAB3B, and TNC) in GFs and CAFs. CCL2, might be an alternative target for treating both epithelial RAB3B, and TNC have known CAF markers in gene set cancers and sarcomas. enrichment analyses (GSEA; MISHRA_CAF_UP)19. The CAFs originate from various cell types, including resi- expression levels of CCL2, RAB3B, and TNC were also dent fibroblasts, epithelial cells, and bone marrow-derived increased in CAFs compared to GFs (Fig. 1d; Supple- mesenchymal stem cells. Cytokines, chemokines, and mentary Fig. S1c). CM from GIST-T1 cells and CAFs growth factors can enhance their transition into CAFs in resulted in increased mRNA expression of CCL2, RAB3B, – tumor stroma13 15. Transforming growth factor-β1 (TGF- and TNC in GFs (Fig. 1e), suggesting that CM of GIST β1), a ligand of the TGF-β receptor complex, plays and CAFs can promote a GF-to-CAF transition. important roles in many biological processes by activating To further investigate the functional role of PDGFC TGF-β signaling, which promotes tumorigenesis and secreted from CAFs in GIST, a phosphokinase array was – metastasis16 18. Moreover, normal fibroblasts derived probed to compare CM from CAFscramble (scr), from bone marrow-derived mesenchymal stem cells in CAFshPDGFC #1, and CAFshPDGFC #2 in GIST-T1. pancreatic cancer stroma have been reported to transition Notably, CAFscr CM significantly activated several into CAFs in response to TGF-β signaling, which enhances phosphokinase proteins, including JNK1/2/3, AKT, FAK, tumor growth and invasion13. However, little is known PRAS40, c-Jun, and STAT3, while these effects were regarding the role of TGF-β signaling in the stroma of abrogated by loss of PDGFC (Fig. 1f, g). In addition, as sarcomas. shown in supplementary Fig. S1d, CAFs markedly induced Here, we present a detailed mechanism for the transi- phosphorylation of PDGFRA and PDGFRB, while this tion of gastric fibroblasts (GFs) into CAFs in GIST, and effect was decreased by the loss of PDGFC. Because how this transition promotes tumor migration and PDGFC is not known to activate PDGFR-β/β, these metastasis. First, TGF-β1 secretion from GISTs and CAFs results suggested that CAF-mediated PDGFC secretion enhances a transition from normal resident fibroblast to affects both PDGFR-α/α homo-dimerization and PDGFR- CAFs. In turn, these ligand-dependent events lead to α/β hetero-dimerization, supporting our previous finding increased tumor motility. Overall, this study suggests that that CAF-mediated PDGFC secretion enhances GIST the TGF-β1-mediated GF-to-CAF transition in the TME progression and metastasis. Together, these data plays an important role in sarcoma migration and demonstrated that PDGFC could be a CAF marker that metastasis, implying that this paracrine network might be modulates an aggressive phenotype of GIST cells. a potential target for treating sarcomas. FGFs are overexpressed in CAFs Results Next, we performed RNA sequencing analyses (RNA- CAFs and GIST cells induce a GF-to-CAF transition seq) using GIST-T1 cells and CAFs to characterize the Recently, we discovered that there are CAFs in GIST CAFs (GSE143547). Interestingly, RNA-seq revealed that (Supplementary Fig. S1a), and platelet-derived growth genes encoding various fibroblast growth factors (FGFs) factor C (PDGFC) secreted from CAFs promotes tumor were overexpressed in CAFs (Fig. 2a). These genes are progression and metastasis via a paracrine PDGFC- known to be upregulated in many cancers, including PDGFRA-SLUG axis12. Since the CAFs were isolated prostate, breast, and colon cancer, as well as are asso- from gastric GIST, we obtained a commercially available ciated with poor survival by promoting tumor growth, – normal GF line to compare with CAFs. To understand the metastasis, and angiogenesis20 22. Thus, we next eval- differences between resident GFs and CAFs isolated from uated the expression of these FGF genes by qPCR. Among human GIST, we compared the morphology and mRNA them, CAFs expressed more FGF1, FGF5, and FGF9 expression of PDGFC, as a functional biomarker of CAFs, expression than GFs (Fig. 2b). In addition, these FGF- in these cells. We observed that CAFs had a more stellate encoding genes were significantly increased in GFs with morphology and higher expression of PDGFC as com- the application of CM from either GIST-T1 cells or CAFs pared to GFs (Fig. 1a, b). In addition, CAFs from GISTs (Fig. 2c), indicating that the expression of these genes with different mutations (i.e., KIT or PDGFRA) and dis- might influence the GIST phenotype. tinct locations (i.e., stomach and rectum) were isolated and characterized. The quantitative polymerase chain reaction TGF-β1 is highly expressed in GISTs and CAFs (qPCR) analysis showed that PDGFC mRNA was highly TGF-β signaling contributes to various biological pro- expressed in all CAF lines (Supplementary Fig. S1b). cesses that promote tumorigenesis and metastasis16,17.In We then cultured the GFs with either GIST-T1 or CAF pancreatic cancer stroma, for example, normal fibroblasts conditioned media (CM). Both CM increased mRNA convert into CAFs via TGF-β signaling, and this process is expression of PDGFC in GFs (Fig. 1c). In addition, we associated with tumor growth and invasion13.OurRNA-seq Oncogenesis Yoon et al. Oncogenesis (2021) 10:13 Page 3 of 12 Fig. 1 (See legend on next page.) Oncogenesis Yoon et al. Oncogenesis (2021) 10:13 Page 4 of 12 (see figure on previous page) Fig.
Recommended publications
  • ARTICLES Fibroblast Growth Factors 1, 2, 17, and 19 Are The
    0031-3998/07/6103-0267 PEDIATRIC RESEARCH Vol. 61, No. 3, 2007 Copyright © 2007 International Pediatric Research Foundation, Inc. Printed in U.S.A. ARTICLES Fibroblast Growth Factors 1, 2, 17, and 19 Are the Predominant FGF Ligands Expressed in Human Fetal Growth Plate Cartilage PAVEL KREJCI, DEBORAH KRAKOW, PERTCHOUI B. MEKIKIAN, AND WILLIAM R. WILCOX Medical Genetics Institute [P.K., D.K., P.B.M., W.R.W.], Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Obstetrics and Gynecology [D.K.] and Department of Pediatrics [W.R.W.], UCLA School of Medicine, Los Angeles, California 90095 ABSTRACT: Fibroblast growth factors (FGF) regulate bone growth, (G380R) or TD (K650E) mutations (4–6). When expressed at but their expression in human cartilage is unclear. Here, we deter- physiologic levels, FGFR3-G380R required, like its wild-type mined the expression of entire FGF family in human fetal growth counterpart, ligand for activation (7). Similarly, in vitro cul- plate cartilage. Using reverse transcriptase PCR, the transcripts for tivated human TD chondrocytes as well as chondrocytes FGF1, 2, 5, 8–14, 16–19, and 21 were found. However, only FGF1, isolated from Fgfr3-K644M mice had an identical time course 2, 17, and 19 were detectable at the protein level. By immunohisto- of Fgfr3 activation compared with wild-type chondrocytes and chemistry, FGF17 and 19 were uniformly expressed within the showed no receptor activation in the absence of ligand (8,9). growth plate. In contrast, FGF1 was found only in proliferating and hypertrophic chondrocytes whereas FGF2 localized predominantly to Despite the importance of the FGF ligand for activation of the resting and proliferating cartilage.
    [Show full text]
  • Role and Regulation of Pdgfra Signaling in Liver Development and Regeneration
    The American Journal of Pathology, Vol. 182, No. 5, May 2013 ajp.amjpathol.org GROWTH FACTORS, CYTOKINES, AND CELL CYCLE MOLECULES Role and Regulation of PDGFRa Signaling in Liver Development and Regeneration Prince K. Awuah,* Kari N. Nejak-Bowen,* and Satdarshan P.S. Monga*y From the Division of Experimental Pathology,* Department of Pathology, and the Department of Medicine,y University of Pittsburgh, Pittsburgh, Pennsylvania Accepted for publication January 22, 2013. Aberrant platelet-derived growth factor receptor-a (PDGFRa) signaling is evident in a subset of hepato- cellular cancers (HCCs). However, its role and regulation in hepatic physiology remains elusive. In the Address correspondence to a fi Satdarshan P.S. Monga, M.D., current study, we examined PDGFR signaling in liver development and regeneration. We identi ed a a Divisions of Experimental notable PDGFR activation in hepatic morphogenesis that, when interrupted by PDGFR -blocking anti- Pathology, Pathology and body, led to decreased hepatoblast proliferation and survival in embryonic liver cultures. We also identified Medicine, University of Pitts- temporal PDGFRa overexpression, which is regulated by epidermal growth factor (EGF) and tumor necrosis burgh School of Medicine, 200 factor a, and its activation at 3 to 24 hours after partial hepatectomy. Through generation of hepatocyte- Lothrop St., S-422 BST, Pitts- specific PDGFRA knockout (KO) mice that lack an overt phenotype, we show that absent PDGFRa burgh, PA 15261. E-mail: compromises extracelluar signal-regulated kinases and AKT activation 3 hours after partial hepatectomy, [email protected]. which, however, is alleviated by temporal compensatory increases in the EGF receptor (EGFR) and the hepatocyte growth factor receptor (Met) expression and activation along with rebound activation of extracellular signal-regulated kinases and AKT at 24 hours.
    [Show full text]
  • Instability Restricts Signaling of Multiple Fibroblast Growth Factors
    Cell. Mol. Life Sci. DOI 10.1007/s00018-015-1856-8 Cellular and Molecular Life Sciences RESEARCH ARTICLE Instability restricts signaling of multiple fibroblast growth factors Marcela Buchtova • Radka Chaloupkova • Malgorzata Zakrzewska • Iva Vesela • Petra Cela • Jana Barathova • Iva Gudernova • Renata Zajickova • Lukas Trantirek • Jorge Martin • Michal Kostas • Jacek Otlewski • Jiri Damborsky • Alois Kozubik • Antoni Wiedlocha • Pavel Krejci Received: 18 June 2014 / Revised: 7 February 2015 / Accepted: 9 February 2015 Ó Springer Basel 2015 Abstract Fibroblast growth factors (FGFs) deliver ex- failure to activate FGF receptor signal transduction over tracellular signals that govern many developmental and long periods of time, and influence specific cell behavior regenerative processes, but the mechanisms regulating FGF in vitro and in vivo. Stabilization via exogenous heparin signaling remain incompletely understood. Here, we ex- binding, introduction of stabilizing mutations or lowering plored the relationship between intrinsic stability of FGF the cell cultivation temperature rescues signaling of un- proteins and their biological activity for all 18 members of stable FGFs. Thus, the intrinsic ligand instability is an the FGF family. We report that FGF1, FGF3, FGF4, FGF6, important elementary level of regulation in the FGF sig- FGF8, FGF9, FGF10, FGF16, FGF17, FGF18, FGF20, and naling system. FGF22 exist as unstable proteins, which are rapidly de- graded in cell cultivation media. Biological activity of Keywords Fibroblast growth factor Á FGF Á Unstable Á FGF1, FGF3, FGF4, FGF6, FGF8, FGF10, FGF16, FGF17, Proteoglycan Á Regulation and FGF20 is limited by their instability, manifesting as Electronic supplementary material The online version of this article (doi:10.1007/s00018-015-1856-8) contains supplementary material, which is available to authorized users.
    [Show full text]
  • Alternative Splicing Generates an Isoform of the Human Sef Gene with Altered Subcellular Localization and Specificity
    Alternative splicing generates an isoform of the human Sef gene with altered subcellular localization and specificity Ella Preger*, Inbal Ziv*, Ariel Shabtay*, Ifat Sher*, Michael Tsang†, Igor B. Dawid†, Yael Altuvia‡, and Dina Ron*§ *Department of Biology, Technion–Israel Institute of Technology, Haifa 32000, Israel; †Laboratory of Molecular Genetics, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892; and ‡Department of Molecular Genetics and Biotechnology, Hebrew University–Hadassah Medical School, Jerusalem 91120, Israel Contributed by Igor B. Dawid, December 1, 2003 Receptor tyrosine kinases (RTKs) control a multitude of biological FGFs comprise a family of 22 structurally related polypeptide processes and are therefore subjected to multiple levels of regu- mitogens that control cell proliferation, differentiation, survival, lation. Negative feedback is one of the mechanisms that provide an and migration and play a key role in embryonic patterning effective means to control RTK-mediated signaling. Sef has re- (14–16). They signal via binding and activation of a family of cently been identified as a specific antagonist of fibroblast growth cell-surface tyrosine kinase receptors designated FGF receptors factor (FGF) signaling in zebrafish and subsequently in mouse and 1–4 (FGFR1–FGFR4) (17–20). Activated receptors trigger sev- human. Sef encodes a putative type I transmembrane protein that eral signal transduction cascades including the Ras͞MAPK and antagonizes the Ras͞mitogen-activated protein kinase pathway in the PI3-kinase pathway (15, 21). Depending on the cell type, all three species. Mouse Sef was also shown to inhibit the phos- FGF can also activate other MAPK pathways, such that leading phatidylinositol 3-kinase pathway.
    [Show full text]
  • PDGF-C and PDGF-D Signaling in Vascular Diseases and Animal Models
    Molecular Aspects of Medicine 62 (2018) 1e11 Contents lists available at ScienceDirect Molecular Aspects of Medicine journal homepage: www.elsevier.com/locate/mam PDGF-C and PDGF-D signaling in vascular diseases and animal models * Erika Folestad a, Anne Kunath b, Dick Wågsater€ b, a Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden b Division of Drug Research, Department of Medical and Health Sciences, Linkoping€ University, Linkoping,€ Sweden article info abstract Article history: Members of the platelet-derived growth factor (PDGF) family are well known to be involved in different Received 31 August 2017 pathological conditions. The cellular and molecular mechanisms induced by the PDGF signaling have Received in revised form been well studied. Nevertheless, there is much more to discover about their functions and some 14 November 2017 important questions to be answered. This review summarizes the known roles of two of the PDGFs, Accepted 22 January 2018 PDGF-C and PDGF-D, in vascular diseases. There are clear implications for these growth factors in several Available online 14 February 2018 vascular diseases, such as atherosclerosis and stroke. The PDGF receptors are broadly expressed in the cardiovascular system in cells such as fibroblasts, smooth muscle cells and pericytes. Altered expression Keywords: Aneurysm of the receptors and the ligands have been found in various cardiovascular diseases and current studies fi Atherosclerosis have shown important implications of PDGF-C and PDGF-D signaling in brosis, neovascularization, Growth factor atherosclerosis and restenosis. Myocardial infarction © 2018 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND Smooth muscle cells license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
    [Show full text]
  • TABLE S1 Complete Overview of Protocols for Induction of Pscs Into
    TABLE S1 Complete overview of protocols for induction of PSCs into renal lineages Ref 2D/ Cell type Protocol Days Growth factors Outcome Other analyses # 3D hiPSC, hESC, Collagen type I tx murine epidydymal fat pads,ex vivo 54 2D 8 Y27632, AA, CHIR, BMP7 IM miPSC, mESC Matrigel with murine fetal kidney hiPSC, hESC, Suspension,han tx murine epidydymal fat pads,ex vivo 54 2D 20 AA, CHIR, BMP7 IM miPSC, mESC ging drop with murine fetal kidney tx murine epidydymal fat pads,ex vivo 55 hiPSC, hESC Matrigel 2D 14 CHIR, TTNPB/AM580, Y27632 IM with murine fetal kidney Injury, tx murine epidydymal fat 57 hiPSC Suspension 2D 28 AA, CHIR, BMP7, TGF-β1, TTNPB, DMH1 NP pads,spinal cord assay Matrigel,membr 58 mNPC, hESC 3D 7 BPM7, FGF9, Heparin, Y27632, CHIR, LDN, BMP4, IGF1, IGF2 NP Clonal expansion ane filter iMatrix,spinal 59 hiPSC 3D 10 LIF, Y27632, FGF2/FGF9, TGF-α, DAPT, CHIR, BMP7 NP Clonal expansion cord assay iMatrix,spinal 59 murine NP 3D 8-19 LIF, Y27632, FGF2/FGF9, TGF- α, DAPT, CHIR, BMP7 NP Clonal expansion cord assay murine NP, Suspension, 60 3D 7-19 BMP7, FGF2, Heparin, Y27632, LIF NP Nephrotoxicity, injury model human NP membrane filter Suspension, Contractility and permeability assay, ex 61 hiPSC 2D 10 AA, BMP7, RA Pod gelatin vivo with murine fetal kidney Matrigel. 62 hiPSC, hESC fibronectin, 2D < 50 FGF2, AA, WNT3A, BMP4, BMP7, RA, FGF2, HGF or RA + VITD3 Pod collagen type I Matrigel, Contractility and uptake assay,ex vivo 63 hiPSC 2D 13 Y27632, CP21R7, BMP4, RA, BMP7, FGF9, VITD3 Pod collagen type I with murine fetal kidney Collagen
    [Show full text]
  • A Novel Fibroblast Growth Factor 1 Variant Reverses Nonalcoholic Fatty Liver Disease in Type 2 Diabetes
    University of Louisville ThinkIR: The University of Louisville's Institutional Repository Electronic Theses and Dissertations 8-2018 A novel fibroblast growth factor 1 variant reverses nonalcoholic fatty liver disease in type 2 diabetes. Qian Lin University of Louisville Follow this and additional works at: https://ir.library.louisville.edu/etd Part of the Endocrine System Diseases Commons, and the Pharmacology Commons Recommended Citation Lin, Qian, "A novel fibroblast growth factor 1 variant reverses nonalcoholic fatty liver disease in type 2 diabetes." (2018). Electronic Theses and Dissertations. Paper 3016. https://doi.org/10.18297/etd/3016 This Doctoral Dissertation is brought to you for free and open access by ThinkIR: The University of Louisville's Institutional Repository. It has been accepted for inclusion in Electronic Theses and Dissertations by an authorized administrator of ThinkIR: The University of Louisville's Institutional Repository. This title appears here courtesy of the author, who has retained all other copyrights. For more information, please contact [email protected]. A NOVEL FIBROBLAST GROWTH FACTOR 1 VARIANT REVERSES NONALCOHOLIC FATTY LIVER DISEASE IN TYPE 2 DIABETES BY Qian Lin M.S. at Wenzhou Medical University, 2016 A Dissertation Submitted to the Faculty of the School of Medicine of the University of Louisville for the Degree of Doctor of Philosophy in Pharmacology and Toxicology Department of Pharmacology and Toxicology University of Louisville Louisville, Kentucky August 2018 A NOVEL FIBROBLAST GROWTH FACTOR 1 VARIANT REVERSES NONALCOHOLIC FATTY LIVER DISEASE IN TYPE 2 DIABETES BY Qian Lin Dissertation Approved on August 2, 2018 Dissertation Committee: Dr. Yi Tan Dr. Paul Epstein Dr.
    [Show full text]
  • The Roles of Fgfs in the Early Development of Vertebrate Limbs
    Downloaded from genesdev.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW The roles of FGFs in the early development of vertebrate limbs Gail R. Martin1 Department of Anatomy and Program in Developmental Biology, School of Medicine, University of California at San Francisco, San Francisco, California 94143–0452 USA ‘‘Fibroblast growth factor’’ (FGF) was first identified 25 tion of two closely related proteins—acidic FGF and ba- years ago as a mitogenic activity in pituitary extracts sic FGF (now designated FGF1 and FGF2, respectively). (Armelin 1973; Gospodarowicz 1974). This modest ob- With the advent of gene isolation techniques it became servation subsequently led to the identification of a large apparent that the Fgf1 and Fgf2 genes are members of a family of proteins that affect cell proliferation, differen- large family, now known to be comprised of at least 17 tiation, survival, and motility (for review, see Basilico genes, Fgf1–Fgf17, in mammals (see Coulier et al. 1997; and Moscatelli 1992; Baird 1994). Recently, evidence has McWhirter et al. 1997; Hoshikawa et al. 1998; Miyake been accumulating that specific members of the FGF 1998). At least five of these genes are expressed in the family function as key intercellular signaling molecules developing limb (see Table 1). The proteins encoded by in embryogenesis (for review, see Goldfarb 1996). Indeed, the 17 different FGF genes range from 155 to 268 amino it may be no exaggeration to say that, in conjunction acid residues in length, and each contains a conserved with the members of a small number of other signaling ‘‘core’’ sequence of ∼120 amino acids that confers a com- molecule families [including WNT (Parr and McMahon mon tertiary structure and the ability to bind heparin or 1994), Hedgehog (HH) (Hammerschmidt et al.
    [Show full text]
  • The FGF/FGFR System in Breast Cancer: Oncogenic Features and Therapeutic Perspectives
    cancers Review The FGF/FGFR System in Breast Cancer: Oncogenic Features and Therapeutic Perspectives Maria Francesca Santolla and Marcello Maggiolini * Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; [email protected] * Correspondence: [email protected] or [email protected] Received: 8 September 2020; Accepted: 16 October 2020; Published: 18 October 2020 Simple Summary: The fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) system represents an emerging therapeutic target in breast cancer. Here, we discussed previous studies dealing with FGFR molecular aberrations, the alterations in the FGF/FGFR signaling across the different subtypes of breast cancer, the functional interplay between the FGF/FGFR axis and important components of the breast microenvironment, the therapeutic usefulness of FGF/FGFR inhibitors for the treatment of breast cancer. Abstract: One of the major challenges in the treatment of breast cancer is the heterogeneous nature of the disease. With multiple subtypes of breast cancer identified, there is an unmet clinical need for the development of therapies particularly for the less tractable subtypes. Several transduction mechanisms are involved in the progression of breast cancer, therefore making the assessment of the molecular landscape that characterizes each patient intricate. Over the last decade, numerous studies have focused on the development of tyrosine kinase inhibitors (TKIs) to target the main pathways dysregulated in breast cancer, however their effectiveness is often limited either by resistance to treatments or the appearance of adverse effects. In this context, the fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) system represents an emerging transduction pathway and therapeutic target to be fully investigated among the diverse anti-cancer settings in breast cancer.
    [Show full text]
  • Upregulated Expression of FGF13/FHF2 Mediates Resistance To
    OPEN Upregulated expression of FGF13/FHF2 SUBJECT AREAS: mediates resistance to platinum drugs in CANCER THERAPEUTIC RESISTANCE cervical cancer cells CANCER PREVENTION Tomoko Okada1, Kazuhiro Murata1,2, Ryoma Hirose1,3, Chie Matsuda1, Tsunehiko Komatsu2, STRESS SIGNALLING Masahiko Ikekita3, Miyako Nakawatari4, Fumiaki Nakayama4, Masaru Wakatsuki5, Tatsuya Ohno5,6, CHEMOTHERAPY Shingo Kato5,7, Takashi Imai4 & Toru Imamura1,3 Received 1Signaling Molecules Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and 27 June 2013 2 Technology (AIST), Tsukuba, Ibaraki 305-8566, Japan, Division of Hematology, 3rd Department of Internal Medicine, Teikyo 3 Accepted University Chiba Medical Center, Ichihara, Chiba 299-0111, Japan, Department of Applied Biological Science, Faculty of Science 4 18 September 2013 and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan, Advanced Radiation Biology Research Program, National Institute of Radiological Sciences, Chiba, Chiba 263-8555, Japan, 5Research Center Hospital for Charged Particle Published Therapy, National Institute of Radiological Sciences, Chiba, Chiba 263-8555, Japan, 6Gunma University Graduate School of 11 October 2013 Medicine, Maebashi, Gunma 371-8511, Japan, 7Saitama Medical University International Medical Center, Hidaka, Saitama 350- 1298, Japan. Correspondence and Cancer cells often develop drug resistance. In cisplatin-resistant HeLa cisR cells, fibroblast growth factor 13 requests for materials (FGF13/FHF2) gene and protein expression was strongly upregulated, and intracellular platinum should be addressed to concentrations were kept low. When the FGF13 expression was suppressed, both the cells’ resistance to T.Imamura (imamura- platinum drugs and their ability to keep intracellular platinum low were abolished. Overexpression of [email protected]) FGF13 in parent cells led to greater resistance to cisplatin and reductions in the intracellular platinum concentration.
    [Show full text]
  • Ep 3217179 A1
    (19) TZZ¥ ___T (11) EP 3 217 179 A1 (12) EUROPEAN PATENT APPLICATION (43) Date of publication: (51) Int Cl.: 13.09.2017 Bulletin 2017/37 G01N 33/68 (2006.01) (21) Application number: 17167637.2 (22) Date of filing: 02.10.2013 (84) Designated Contracting States: • LIU, Xinjun AL AT BE BG CH CY CZ DE DK EE ES FI FR GB San Diego, CA 92130 (US) GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO • HAUENSTEIN, Scott PL PT RO RS SE SI SK SM TR San Diego, CA 92130 (US) • KIRKLAND, Richard (30) Priority: 05.10.2012 US 201261710491 P San Diego, CA 92111 (US) 17.05.2013 US 201361824959 P (74) Representative: Krishnan, Sri (62) Document number(s) of the earlier application(s) in Nestec S.A. accordance with Art. 76 EPC: Centre de Recherche Nestlé 13779638.9 / 2 904 405 Vers-chez-les-Blanc Case Postale 44 (71) Applicant: Nestec S.A. 1000 Lausanne 26 (CH) 1800 Vevey (CH) Remarks: (72) Inventors: This application was filed on 21-04-2017 as a • SINGH, Sharat divisional application to the application mentioned Rancho Santa Fe, CA 92127 (US) under INID code 62. (54) METHODS FOR PREDICTING AND MONITORING MUCOSAL HEALING (57) The present invention provides methods for pre- an individual with a disease such as IBD. Information on dicting the likelihood of mucosal healing in an individual mucosal healing status derived from the use of the with a disease such as inflammatory bowel disease present invention can also aid in optimizing therapy (IBD).
    [Show full text]
  • Type I and II Interferon Are Associated with High Expression of the Hippo Pathway Family Members
    Cel al & lul ic ar in I l m C m f o u n l a o l n o Journal of Clinical & Cellular r g u y o J ISSN: 2155-9899 Immunology Research Article Type I and II Interferon are Associated with High Expression of the Hippo Pathway Family Members Bianca Sciescia1*, Raquel Tognon2, Natalia de Souza Nunes1, Tathiane Maistro Malta1, Fabiani Gai Frantz1, Fabiola Attie de Castro1, Maira da Costa Cacemiro1 1Department of Clinical, Toxicological and Bromatological Analysis, University of Sao Paulo - USP, Ribeirao Preto - SP, Brazil; 2Department of Pharmacy, Federal University of Juiz de Fora, Campus Governador Valadares, Governador Valadares - MG, Brazil ABSTRACT The Hippo pathway plays a regulatory role on inflammation and cell death and proliferation. Here we described a relationship between Hippo pathway components and inflammation in healthy subjects. The plasma levels of cytokines and chemokines were used to define their inflammatory profile and classify them as normal, high and low producers of cytokines. Leukocytes from healthy subjects with inflammatory profile expressed the highest levels of MSTS1/MST2, SAV1, LATS1/LATS2, MOB1A/MOB1B and YAP genes. The group that overexpressed Hippo pathway-related genes secreted more IFN-ϒ and IFN-α2. Keywords: Hippo pathway; Inflammatory process; Healthy subjects; Cytokines; Chemokines ABBREVATIONS: LATS1/LATS2 kinases (large tumor suppressor kinase 1/2) and MOB kinase activator IL: Interleukin; IFN: Interferon; LATS1/LATS2: Large tumor their adapter proteins MOB1A/MOB1B ( 1A/1B yes-associated suppressor kinase 1/2; MCP-1: Monocyte chemoattractant ), and the transcription coactivators YAP ( protein tafazzin protein protein-1; MIP: Macrophage inflammatory protein; MOB1A/ ) and TAZ ( ).
    [Show full text]