1
|
Hammoudeh SM, Ng Y, Wei BR, Madsen TD, Yadav MP, Simpson RM, Weigert R, Randazzo PA. Tongue orthotopic xenografts to study fusion-negative rhabdomyosarcoma invasion and metastasis in live animals. CELL REPORTS METHODS 2024; 4:100802. [PMID: 38964316 PMCID: PMC11294838 DOI: 10.1016/j.crmeth.2024.100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/24/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024]
Abstract
PAX3/7 fusion-negative rhabdomyosarcoma (FN-RMS) is a childhood mesodermal lineage malignancy with a poor prognosis for metastatic or relapsed cases. Limited understanding of advanced FN-RMS is partially attributed to the absence of sequential invasion and dissemination events and the challenge in studying cell behavior, using, for example, non-invasive intravital microscopy (IVM), in currently used xenograft models. Here, we developed an orthotopic tongue xenograft model of FN-RMS to study cell behavior and the molecular basis of invasion and metastasis using IVM. FN-RMS cells are retained in the tongue and invade locally into muscle mysial spaces and vascular lumen, with evidence of hematogenous dissemination to the lungs and lymphatic dissemination to lymph nodes. Using IVM of tongue xenografts reveals shifts in cellular phenotype, migration to blood and lymphatic vessels, and lymphatic intravasation. Insight from this model into tumor invasion and metastasis at the tissue, cellular, and subcellular level can guide new therapeutic avenues for advanced FN-RMS.
Collapse
Affiliation(s)
- Sarah M Hammoudeh
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Yeap Ng
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; CCR-Intravital Microscopy Core, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Thomas D Madsen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Copenhagen Center for Glycomics, University of Copenhagen, Department for Cellular and Molecular Medicine, Copenhagen, Denmark
| | - Mukesh P Yadav
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - R Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; CCR-Intravital Microscopy Core, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
2
|
Hammoudeh SM, Ng Y, Wei BR, Madsen TD, Simpson RM, Weigert R, Randazzo PA. Fusion-negative rhabdomyosarcoma orthotopic tongue xenografts for study of invasion, intravasation and metastasis in live animals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558858. [PMID: 38076999 PMCID: PMC10705524 DOI: 10.1101/2023.09.21.558858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
PAX3/7 Fusion-negative rhabdomyosarcoma (FN-RMS) is a childhood mesodermal lineage malignancy with a poor prognosis for metastatic or relapsed cases. Towards achieving a more complete understanding of advanced FN-RMS, we developed an orthotopic tongue xenograft model for studies of molecular basis of FN-RMS invasion and metastasis. The behavior of FN-RMS cells injected into murine tongue was examined using in vivo bioluminescence imaging, non-invasive intravital microscopy (IVM), and histopathology and compared to the prevailing hindlimb intramuscular and subcutaneous xenografts. FN-RMS cells were retained in the tongue and invaded locally into muscle mysial spaces and vascular lumen. While evidence of hematogenous dissemination to the lungs occurred in tongue and intramuscular xenografts, evidence of local invasion and lymphatic dissemination to lymph nodes only occurred in tongue xenografts. IVM and RNA-seq of tongue xenografts reveal shifts in cellular phenotype and differentiation state in tongue xenografts. IVM also shows homing to blood and lymphatic vessels, lymphatic intravasation, and dynamic membrane protrusions. Based on these findings, the tongue orthotopic xenograft of FN-RMS is a valuable model for tumor progression studies at the tissue, cellular and subcellular levels providing insight into kinetics and molecular bases of tumor invasion and metastasis and, hence, new therapeutic avenues for advanced FN-RMS.
Collapse
Affiliation(s)
- Sarah M Hammoudeh
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Yeap Ng
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
- CCR-Intravital Microscopy Core, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Thomas D Madsen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
- Copenhagen Center for Glycomics, University of Copenhagen, Department for Cellular and Molecular Medicine; Copenhagen, Denmark
| | - R Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
- CCR-Intravital Microscopy Core, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Chen J, Li F, Gu J, Zhang X, Bartoli M, Domena JB, Zhou Y, Zhang W, Paulino V, C L B Ferreira B, Michael Brejcha N, Luo L, Arduino C, Verde F, Zhang F, Zhang F, Tagliaferro A, Olivier JH, Zhang Y, Leblanc RM. Cancer cells inhibition by cationic carbon dots targeting the cellular nucleus. J Colloid Interface Sci 2023; 637:193-206. [PMID: 36701865 PMCID: PMC9957951 DOI: 10.1016/j.jcis.2023.01.086] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/31/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Nucleus targeting is tremendously important in cancer therapy. Cationic carbon dots (CCDs) are potential nanoparticles which might enter cells and penetrate nuclear membranes. Although some CCDs have been investigated in nucleus targeting and applied in nuclear imaging, the CCDs derived from drugs, that are able to target the nucleus, bind with DNA and inhibit the growth of cancer cells have not been reported. In this project, 1, 2, 4, 5-benzenetetramine (Y15, a focal adhesion kinase inhibitor) derived cationic carbon dots (Y15-CDs) were prepared via a hydrothermal approach utilizing Y15, folic acid and 1,2-ethylenediamine as precursors. Based on the structural, optical, and morphologic characterizations, Y15-CDs possess rich amine groups and nitrogen in structure, an excitation-dependent photoluminescence emission, and a small particle size of 2 to 4 nm. The DNA binding experiments conducted through agarose gel electrophoresis, UV-vis absorption, fluorescence emission, and circular dichroism spectroscopies, prove that Y15-CDs might bind with DNA via electrostatic interactions and partially intercalative binding modes. In addition, the cell imaging and cytotoxicity studies in human foreskin fibroblasts (HFF), prostate cancer (PC3) and osteosarcoma cells (U2OS) indicate the nucleus targeting and anticancer abilities of Y15-CDs. Most interestingly, Y15-CDs exhibit a higher cytotoxicity to cancer cells (PC3 and U2OS) than to normal cells (HFF), inferring that Y15-CDs might be potentially applied in cancer therapy.
Collapse
Affiliation(s)
- Jiuyan Chen
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | - Fang Li
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Jun Gu
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Xiao Zhang
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | - Mattia Bartoli
- Department of Applied Science and Technology, Politecnico di Torino, Italy
| | - Justin B Domena
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | - Yiqun Zhou
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA; C-Dots, LLC, Miami, FL 33136, USA
| | - Wei Zhang
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | - Victor Paulino
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | | | - Nicholas Michael Brejcha
- Department of Biochemistry and Molecular Biology, University of Miami, Coral Gables, FL 33146, USA
| | - Liang Luo
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Chiara Arduino
- Department of Applied Science and Technology, Politecnico di Torino, Italy
| | - Fulvia Verde
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Fangliang Zhang
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Fuwu Zhang
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | | | | | - Yanbin Zhang
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, FL 33136, USA.
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA.
| |
Collapse
|
4
|
Targeting the FAK-Src Complex in Desmoplastic Small Round Cell Tumors, Ewing Sarcoma, and Rhabdomyosarcoma. Sarcoma 2022; 2022:3089424. [PMID: 35655525 PMCID: PMC9153931 DOI: 10.1155/2022/3089424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/31/2022] [Indexed: 11/21/2022] Open
Abstract
Desmoplastic small round cell tumors (DSRCTs), Ewing sarcoma (ES), and alveolar and embryonal rhabdomyosarcoma (ARMS and ERMS) are malignant sarcomas typically occurring at young age, with a poor prognosis in the metastatic setting. New treatment options are necessary. Src family kinase inhibitor dasatinib single-agent treatment has been investigated in a phase 2 study in patients with advanced sarcomas including ES and RMS but failed as a single agent in these subtypes. Since previous studies demonstrated high FAK and Src activities in RMS and ES tissue and cell lines, and dasatinib treatment was shown to upregulate activated FAK, we hypothesized that FAK-Src combination treatment could potentially be an interesting treatment option for these tumor types. We examined the effects of targeting the FAK-Src complex by addressing (p)FAK and (p)Src expressions in tumor sections of DSRCT (n = 13), ES (n = 68), ARMS (n = 21), and ERMS (n = 39) and by determining the antitumor effects of single and combined treatment with FAK inhibitor defactinib and multikinase (Abl/SFK) inhibitor dasatinib in vitro on cell lines of each subtype. In vivo effects were assessed in DSRCT and ERMS models. Concurrent pFAK and pSrc expressions (H-score >50) were observed in DSRCT (67%), ES (6%), ARMS (35%), and ERMS (19%) samples. Defactinib treatment decreased pFAK expression and reduced cell viability in all subtypes. Dasatinib treatment decreased pSrc expression and cell viability in each subtype. Combination treatment led to a complete reduction in pFAK and pSrc in each cell line and showed enhanced cell viability reduction, drug synergy, DNA damage induction, and a trend toward higher apoptosis induction in DSRCT, ERMS, and ARMS but not in ES cells. These promising in vitro results unfortunately do not translate into promising in vivo results as we did not observe a significant effect on tumor volume in vivo, and the combination did not show superior effects compared to dasatinib single-agent treatment.
Collapse
|
5
|
Pomella S, Cassandri M, Braghini MR, Marampon F, Alisi A, Rota R. New Insights on the Nuclear Functions and Targeting of FAK in Cancer. Int J Mol Sci 2022; 23:ijms23041998. [PMID: 35216114 PMCID: PMC8874710 DOI: 10.3390/ijms23041998] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase over-expressed and activated in both adult and pediatric cancers, where it plays important roles in the regulation of pathogenesis and progression of the malignant phenotype. FAK exerts its functions in cancer by two different ways: a kinase activity in the cytoplasm, mainly dependent on the integrin signaling, and a scaffolding activity into the nucleus by networking with different gene expression regulators. For this reason, FAK has to be considered a target with high therapeutic values. Indeed, evidence suggests that FAK targeting could be effective, either alone or in combination, with other already available treatments. Here, we propose an overview of the novel insights about FAK’s structure and nuclear functions, with a special focus on the recent findings concerning the roles of this protein in cancer. Additionally, we provide a recent update on FAK inhibitors that are currently in clinical trials for patients with cancer, and discuss the challenge and future directions of drug-based anti-FAK targeted therapies.
Collapse
Affiliation(s)
- Silvia Pomella
- Department of Oncohematology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (M.C.)
| | - Matteo Cassandri
- Department of Oncohematology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (M.C.)
- Department of Radiotherapy, Policlinico Umberto I, Sapienza University of Rome, 00185 Rome, Italy;
| | - Maria Rita Braghini
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, Sapienza University of Rome, 00185 Rome, Italy;
| | - Anna Alisi
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
- Correspondence: (A.A.); (R.R.); Tel.: +39-06-68592186 (A.A.); +39-06-68592648 (R.R.)
| | - Rossella Rota
- Department of Oncohematology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (M.C.)
- Correspondence: (A.A.); (R.R.); Tel.: +39-06-68592186 (A.A.); +39-06-68592648 (R.R.)
| |
Collapse
|
6
|
FAK Signaling in Rhabdomyosarcoma. Int J Mol Sci 2020; 21:ijms21228422. [PMID: 33182556 PMCID: PMC7697003 DOI: 10.3390/ijms21228422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/07/2020] [Accepted: 11/08/2020] [Indexed: 01/01/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of children and adolescents. The fusion-positive (FP)-RMS variant expressing chimeric oncoproteins such as PAX3-FOXO1 and PAX7-FOXO1 is at high risk. The fusion negative subgroup, FN-RMS, has a good prognosis when non-metastatic. Despite a multimodal therapeutic approach, FP-RMS and metastatic FN-RMS often show a dismal prognosis with 5-year survival of less than 30%. Therefore, novel targets need to be discovered to develop therapies that halt tumor progression, reducing long-term side effects in young patients. Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that regulates focal contacts at the cellular edges. It plays a role in cell motility, survival, and proliferation in response to integrin and growth factor receptors’ activation. FAK is often dysregulated in cancer, being upregulated and/or overactivated in several adult and pediatric tumor types. In RMS, both in vitro and preclinical studies point to a role of FAK in tumor cell motility/invasion and proliferation, which is inhibited by FAK inhibitors. In this review, we summarize the data on FAK expression and modulation in RMS. Moreover, we give an overview of the approaches to inhibit FAK in both preclinical and clinical cancer settings.
Collapse
|
7
|
Aye JM, Stafman LL, Williams AP, Garner EF, Stewart JE, Anderson JC, Mruthyunjayappa S, Waldrop MG, Goolsby CD, Markert HR, Quinn C, Marayati R, Mroczek-Musulman E, Willey CD, Yoon KJ, Whelan KF, Beierle EA. The effects of focal adhesion kinase and platelet-derived growth factor receptor beta inhibition in a patient-derived xenograft model of primary and metastatic Wilms tumor. Oncotarget 2019; 10:5534-5548. [PMID: 31565187 PMCID: PMC6756857 DOI: 10.18632/oncotarget.27165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/12/2019] [Indexed: 12/01/2022] Open
Abstract
Aggressive therapies for patients with metastatic Wilms tumor (WT) with subsequent severe late effects warrant the search for novel therapies. The role of focal adhesion kinase (FAK), a non-receptor tyrosine kinase important in pediatric solid tumor development and progression, has not been examined in metastatic WT. Using a novel patient-derived xenograft (PDX) of a primary and matched, isogenic, metastatic WT, the hypothesis of the current study was that FAK would contribute to metastatic WT and small molecule inhibition would decrease tumor growth. Immunohistochemical staining, immunoblotting, cell viability and proliferation assays, cell cycle analysis, and cellular motility and attachment-independent growth assays were performed. FAK was present and phosphorylated in both WT PDXs and in the human samples from which they were derived. FAK inhibition decreased cellular survival, proliferation, and cell cycle progression in both PDXs but only significantly decreased migration, invasion, and attachment-independent growth in the primary WT PDX. Kinomic profiling revealed that platelet-derived growth factor receptor beta (PDGFRβ) may be affected by FAK inhibition in WT. Pharmacologic inhibition of FAK and PDGFRβ was synergistic in primary WT PDX cells. These findings broaden the knowledge of metastatic WT and support further investigations on the potential use of FAK and PDGFRβ inhibitors.
Collapse
Affiliation(s)
- Jamie M. Aye
- Department of Pediatrics, Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Laura L. Stafman
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adele P. Williams
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Evan F. Garner
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jerry E. Stewart
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joshua C. Anderson
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Mary G. Waldrop
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caroline D. Goolsby
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hooper R. Markert
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Colin Quinn
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Raoud Marayati
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Christopher D. Willey
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Karina J. Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kimberly F. Whelan
- Department of Pediatrics, Division of Hematology Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | |
Collapse
|
8
|
Codenotti S, Faggi F, Ronca R, Chiodelli P, Grillo E, Guescini M, Megiorni F, Marampon F, Fanzani A. Caveolin-1 enhances metastasis formation in a human model of embryonal rhabdomyosarcoma through Erk signaling cooperation. Cancer Lett 2019; 449:135-144. [DOI: 10.1016/j.canlet.2019.02.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/08/2019] [Accepted: 02/10/2019] [Indexed: 11/15/2022]
|
9
|
Al-Ghabkari A, Qasrawi DO, Alshehri M, Narendran A. Focal adhesion kinase (FAK) phosphorylation is a key regulator of embryonal rhabdomyosarcoma (ERMS) cell viability and migration. J Cancer Res Clin Oncol 2019; 145:1461-1469. [PMID: 31006845 DOI: 10.1007/s00432-019-02913-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma in children. Pathogenesis of RMS is associated with aggressive growth pattern and increased risk of morbidity and mortality. There are two main subtypes or RMS: embryonal and alveolar. The embryonal type is characterized by distinct molecular aberrations, including alterations in the activity of certain protein kinases. Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that plays a vital role in focal adhesion (FA) assembly to promote cytoskeleton dynamics and regulation of cell motility. It is regulated by multiple phosphorylation sites: tyrosine 397, Tyr 576/577, and Tyr 925. Tyrosine 397 is the autophosphorylation site that regulates FAK localization at the cell periphery to facilitate the assembly and formation of the FA complex. The kinase activity of FAK is mediated by the phosphorylation of Tyr 576/577 within the kinase domain activation loop. Aberrations of FAK phosphorylation have been linked to the pathogenesis of different types of cancers. In this regard, pY397 upregulation is linked to increase ERMS cell motility, invasion, and tumorigenesis. METHODS In this study, we have used an established human embryonal muscle rhabdomyosarcoma cell line RD as a model to examine FAK phosphorylation profiles to characterize its role in the pathogenies of RMS. RESULTS Our findings revealed a significant increase of FAK phosphorylation at pY397 in RD cells compared to control cells (hTERT). On the other hand, Tyr 576/577 phosphorylation levels in RD cells displayed a pronounced reduction. Our data showed that Y925 residue exhibited no detectable change. The in vitro analysis showed that the FAK inhibitor, PF-562271 led to G1 cell-cycle arrest induced cell death (IC50, ~ 12 µM) compared to controls. Importantly, immunostaining analyses displayed a noticeable reduction of Y397 phosphorylation following PF-562271 treatment. Our data also showed that PF-562271 suppressed RD cell migration in a dose-dependent manner associated with a reduction in Y397 phosphorylation. CONCLUSIONS The data presented herein indicate that targeting FAK phosphorylation at distinct sites is a promising strategy in future treatment approaches for defined subgroups of rhabdomyosarcoma.
Collapse
Affiliation(s)
- Abdulhameed Al-Ghabkari
- Department of Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada.
| | - Deema O Qasrawi
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Mana Alshehri
- Department of Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Aru Narendran
- Department of Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
| |
Collapse
|
10
|
Namatame N, Tamaki N, Yoshizawa Y, Okamura M, Nishimura Y, Yamazaki K, Tanaka M, Nakamura T, Semba K, Yamori T, Yaguchi SI, Dan S. Antitumor profile of the PI3K inhibitor ZSTK474 in human sarcoma cell lines. Oncotarget 2018; 9:35141-35161. [PMID: 30416685 PMCID: PMC6205545 DOI: 10.18632/oncotarget.26216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/26/2018] [Indexed: 11/25/2022] Open
Abstract
Treatment of patients with advanced sarcoma remains challenging due to lack of effective medicine, with the development of novel drugs being of keen interest. A pan-PI3K inhibitor, ZSTK474, has been evaluated in clinical trials against a range of advanced solid tumors, with clinical benefit shown in sarcoma patients. In the present study, we developed a panel of 14 human sarcoma cell lines and investigated the antitumor effect of 24 anticancer agents including ZSTK474, other PI3K inhibitors, and those clinically used for sarcoma treatment. ZSTK474 exhibited a similar antiproliferative profile to other PI3K inhibitors but was clearly different from the other drugs examined. Indeed, ZSTK474 inhibited PI3K-downstream pathways, in parallel to growth inhibition, in all cell lines examined, showing proof-of-concept of PI3K inhibition. In addition, ZSTK474 induced apoptosis selectively in Ewing's sarcoma (RD-ES and A673), alveolar rhabdomyosarcoma (SJCRH30) and synovial sarcoma (SYO-1, Aska-SS and Yamato-SS) cell lines, all of which harbor chromosomal translocation and resulting oncogenic fusion genes, EWSR1-FLI1, PAX3-FOXO1 and SS18-SSX, respectively. Finally, animal experiments confirmed the antitumor activity of ZSTK474 in vivo, with superior efficacy observed in translocation-positive cells. These results suggest that ZSTK474 could be a promising drug candidate for treating sarcomas, especially those harboring chromosomal translocation.
Collapse
Affiliation(s)
- Nachi Namatame
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,R&D Center, Zenyaku Kogyo Co. Ltd, Tokyo, Japan
| | - Naomi Tamaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuya Yoshizawa
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mutsumi Okamura
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yumiko Nishimura
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kanami Yamazaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Miwa Tanaka
- Division of Carcinogenesis, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takuro Nakamura
- Division of Carcinogenesis, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Takao Yamori
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Present address: Center for Product Evaluation, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Shin-Ichi Yaguchi
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,R&D Center, Zenyaku Kogyo Co. Ltd, Tokyo, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
11
|
Tornin J, Hermida-Prado F, Padda RS, Gonzalez MV, Alvarez-Fernandez C, Rey V, Martinez-Cruzado L, Estupiñan O, Menendez ST, Fernandez-Nevado L, Astudillo A, Rodrigo JP, Lucien F, Kim Y, Leong HS, Garcia-Pedrero JM, Rodriguez R. FUS-CHOP Promotes Invasion in Myxoid Liposarcoma through a SRC/FAK/RHO/ROCK-Dependent Pathway. Neoplasia 2017; 20:44-56. [PMID: 29190494 PMCID: PMC5747526 DOI: 10.1016/j.neo.2017.11.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 01/13/2023] Open
Abstract
Deregulated SRC/FAK signaling leads to enhanced migration and invasion in many types of tumors. In myxoid and round cell liposarcoma (MRCLS), an adipocytic tumor characterized by the expression of the fusion oncogene FUS-CHOP, SRC have been found as one of the most activated kinases. Here we used a cell-of-origin model of MRCLS and an MRCLS cell line to thoroughly characterize the mechanisms of cell invasion induced by FUS-CHOP using in vitro (3D spheroid invasion assays) and in vivo (chicken chorioallantoic membrane model) approaches. FUS-CHOP expression activated SRC-FAK signaling and increased the invasive ability of MRCLS cells. In addition, FAK expression was found to significantly correlate with tumor aggressiveness in sarcoma patient samples. The involvement of SRC/FAK activation in FUS-CHOP-mediated invasion was further confirmed using the SRC inhibitor dasatinib, the specific FAK inhibitor PF-573228, and FAK siRNA. Notably, dasatinib and PF573228 could also efficiently block the invasion of cancer stem cell subpopulations. Downstream of SRC/FAK signaling, we found that FUS-CHOP expression increases the levels of the RHO/ROCK downstream effector phospho-MLC2 (T18/S19) and that this activation was prevented by dasatinib or PF573228. Moreover, the ROCK inhibitor RKI-1447 was able to completely abolish invasion in FUS-CHOP-expressing cells. These data uncover the involvement of SRC/FAK/RHO/ROCK signaling axis in FUS-CHOP-mediated invasion, thus providing a rationale for testing inhibitors of this pathway as potential novel antimetastatic agents for MRCLS treatment.
Collapse
Affiliation(s)
- Juan Tornin
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Francisco Hermida-Prado
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | - Ranjit Singh Padda
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, ON, Canada
| | - M Victoria Gonzalez
- CIBER de Cáncer (CIBERONC), Madrid, Spain; Departamento de Cirugía, Universidad de Oviedo and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | | | - Veronica Rey
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Lucia Martinez-Cruzado
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Oscar Estupiñan
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Sofia T Menendez
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | - Lucia Fernandez-Nevado
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Aurora Astudillo
- Servicio de Anatomía Patológica, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Juan P Rodrigo
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | | | - Yohan Kim
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, ON, Canada; Department of Urology, Mayo Clinic, Rochester, MN
| | - Hon S Leong
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, ON, Canada; Department of Urology, Mayo Clinic, Rochester, MN
| | - Juana Maria Garcia-Pedrero
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain.
| | - Rene Rodriguez
- Hospital Universitario Central de Asturias-Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
12
|
Niba ETE, Yamanaka R, Rani AQM, Awano H, Matsumoto M, Nishio H, Matsuo M. DMD transcripts in CRL-2061 rhabdomyosarcoma cells show high levels of intron retention by intron-specific PCR amplification. Cancer Cell Int 2017; 17:58. [PMID: 28546788 PMCID: PMC5442858 DOI: 10.1186/s12935-017-0428-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The DMD gene encoding dystrophin is mutated in Duchenne muscular dystrophy, a fatal progressive muscle wasting disease. DMD has also been shown to act as a tumor suppressor gene. Rhabdomyosarcoma (RMS) is a mesodermal sarcoma that shares characteristics of skeletal muscle precursors. Products of the DMD gene in RMS have not yet been fully clarified. Here, DMD products were analyzed in CRL-2061 cells established from alveolar RMS. METHODS The 14-kb long DMD cDNA was PCR amplified as 20 separated fragments, as were nine short intron regions. Dystrophin was analyzed by Western blotting using an antibody against the C-terminal region of dystrophin. RESULTS Sixteen of the 20 DMD cDNA fragments could be amplified from CRL-2061 cells as muscle cDNA. Three fragments included aberrant gene products, including one in which exon 71 was omitted and one each with retention of introns 40 and 58. In one fragment, extending from exon 70 to 79, no normally spliced product was obtained. Rather, six alternatively spliced products were identified, including a new product deleting exon 73, with the most abundant product showing deletion of exon 78. Although dystrophin expression was expected in CRL-2061 cells, western blotting of cell lysates showed no evidence of dystrophin, suggesting that translation of full-length DMD mRNA was inhibited by intron retention that generated a premature stop codon. Intron specific PCR amplification of nine short introns, showed retention of introns 40, 58, and 70, which constituted about 60, 25 and 9%, respectively, of the total PCR amplified products. The most abundant DMD transcript contained two abnormalities, intron 40 retention and exon 78 skipping. CONCLUSIONS Intron-specific PCR amplification showed that DMD transcripts contained high levels of introns 40, 58 and 70. Retention of these introns may have been responsible for the lack of dystrophin expression by CRL-2061 cells, thereby abolishing the tumor suppressor activity of dystrophin.
Collapse
Affiliation(s)
- Emma Tabe Eko Niba
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani, Nishi, Kobe, 6512180 Japan
| | - Ryo Yamanaka
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani, Nishi, Kobe, 6512180 Japan
| | - Abdul Qawee Mahyoob Rani
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani, Nishi, Kobe, 6512180 Japan.,Department of Pediatrics, Kobe University Graduate School of Medicine, Chuo, Kobe, 6500017 Japan
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Chuo, Kobe, 6500017 Japan
| | - Masaaki Matsumoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Chuo, Kobe, 6500017 Japan
| | - Hisahide Nishio
- Department of Community Medicine and Social Healthcare Sciences, Kobe University Graduate School of Medicine, Chuo, Kobe, 6500017 Japan
| | - Masafumi Matsuo
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani, Nishi, Kobe, 6512180 Japan
| |
Collapse
|
13
|
Kratimenos P, Koutroulis I, Syriopoulou V, Michailidi C, Delivoria-Papadopoulos M, Klijanienko J, Theocharis S. FAK-Src-paxillin system expression and disease outcome in human neuroblastoma. Pediatr Hematol Oncol 2017; 34:221-230. [PMID: 29040002 DOI: 10.1080/08880018.2017.1360969] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Neuroblastoma (NB) often presents with metastatic disease and poor survival. The need for new prognostic markers remains invaluable. The FAK-Src-Paxillin protein system is associated with aggressive phenotype in adult malignancies but is largely unexplored in pediatric NB. OBJECTIVE To assess FAK-Src-Paxillin protein expression in human NB cell lines and clinical cytology material and to delineate its association with survival. DESIGN/METHODS Western blot and immunohistochemistry were applied for FAK-Src-Paxillin expression in NB cell lines and 23 human cytology specimens, respectively. Protein expression in human clinical samples was correlated with clinicopathological parameters, MYCN amplification and survival. RESULTS FAK, Src and Paxillin proteins are expressed in human NB cells lines, and can be detected in clinical cytology specimens from NB patients, (59%, 32% and 33% respectively). Simultaneous FAK-Src-Paxillin expression was noted in 30% of NB patients. Children with concomitant positivity FAK, Src, and Paxillin tumors, as well as MYCN amplification, had increased mortality compared to those without. CONCLUSIONS FAK-Src-Paxillin system is a marker of unfavorable prognosis for human NB patients but also a promising therapeutic target.
Collapse
Affiliation(s)
- Panagiotis Kratimenos
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Children's National Medical Center , The George Washington University, School of Medicine and Health Sciences , Washington, DC , USA.,b First Department of Pathology , National and Kapodistrian University of Athens, School of Medicine , Athens , Greece
| | - Ioannis Koutroulis
- c Department of Pediatrics, Division of Emergency Medicine, Children's National Medical Center , The George Washington University, School of Medicine and Health Sciences , Washington, DC , USA
| | - Vasiliki Syriopoulou
- f National and Kapodistrian University of Athens , School of Medicine, Children's Hospital of Athens, Department of Pediatrics , Athens , Greece
| | - Christina Michailidi
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Children's National Medical Center , The George Washington University, School of Medicine and Health Sciences , Washington, DC , USA
| | | | | | - Stamatios Theocharis
- a Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Children's National Medical Center , The George Washington University, School of Medicine and Health Sciences , Washington, DC , USA.,d Department of Pathology , Institut Curie , Paris , France
| |
Collapse
|