1
|
Gustafson AL, Durbin AD, Artinger KB, Ford HL. Myogenesis gone awry: the role of developmental pathways in rhabdomyosarcoma. Front Cell Dev Biol 2025; 12:1521523. [PMID: 39902277 PMCID: PMC11788348 DOI: 10.3389/fcell.2024.1521523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Rhabdomyosarcoma is a soft-tissue sarcoma that occurs most frequently in pediatric patients and has poor survival rates in patients with recurrent or metastatic disease. There are two major sub-types of RMS: fusion-positive (FP-RMS) and fusion-negative (FN-RMS); with FP-RMS typically containing chromosomal translocations between the PAX3/7-FOXO1 loci. Regardless of subtype, RMS resembles embryonic skeletal muscle as it expresses the myogenic regulatory factors (MRFs), MYOD1 and MYOG. During normal myogenesis, these developmental transcription factors (TFs) orchestrate the formation of terminally differentiated, striated, and multinucleated skeletal muscle. However, in RMS these TFs become dysregulated such that they enable the sustained properties of malignancy. In FP-RMS, the PAX3/7-FOXO1 chromosomal translocation results in restructured chromatin, altering the binding of many MRFs and driving an oncogenic state. In FN-RMS, re-expression of MRFs, as well as other myogenic TFs, blocks terminal differentiation and holds cells in a proliferative, stem-cell-like state. In this review, we delve into the myogenic transcriptional networks that are dysregulated in and contribute to RMS progression. Advances in understanding the mechanisms through which myogenesis becomes stalled in RMS will lead to new tumor-specific therapies that target these aberrantly expressed developmental transcriptional pathways.
Collapse
Affiliation(s)
- Annika L. Gustafson
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Adam D. Durbin
- Division of Molecular Oncology, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Kristin B. Artinger
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN, United States
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
2
|
Adekeye TE, Teets EM, Tomak EA, Waterman SL, Sprague KA, White A, Coffin ML, Varga SM, Easterbrooks TE, Shepherd SJ, Austin JD, Krivorotko D, Hupper TE, Kelley JB, Amacher SL, Talbot JC. Fast-twitch myofibrils grow in proportion to Mylpf dosage in the zebrafish embryo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613721. [PMID: 39345555 PMCID: PMC11429778 DOI: 10.1101/2024.09.18.613721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Muscle cells become stronger by expanding myofibrils, the chains of sarcomeres that produce contraction. Here we investigate how Mylpf (Myosin Light Chain Phosphorylatable Fast) abundance impacts myofibril assembly in fast-twitch muscle. The two zebrafish Mylpf genes (mylpfa and mylpfb) are exclusively expressed in fast-twitch muscle. We show that these cells initially produce six times more mylpfa mRNA and protein than mylpfb. The combined Mylpf protein dosage is necessary for and proportionate to fast-twitch myofibril growth in the embryo. Fast-twitch myofibrils are severely reduced in the mylpfa -/- mutant, leading to loss of high-speed movement; however, by persistent slow movement this mutant swims as far through time as its wild-type sibling. Although the mylpfb -/- mutant has normal myofibrils, myofibril formation fails entirely in the mylpfa -/- ;mylpfb -/- double mutant, indicating that the two genes are collectively essential to myofibril formation. Fast-twitch myofibril width is restored in the mylpfa -/- mutant by transgenic expression of mylpfa-GFP, mylpfb-GFP, and by human MYLPF-GFP to a degree corresponding linearly with GFP brightness. This correlate is inverted by expression of MYLPF alleles that cause Distal Arthrogryposis, which reduce myofibril size in proportion to protein abundance. These effects indicate that Mylpf dosage controls myofibril growth, impacting embryonic development and lifelong health.
Collapse
Affiliation(s)
- Tayo E Adekeye
- School of Biology and Ecology, the University of Maine, 04469, USA
| | - Emily M Teets
- Molecular Genetics, The Ohio State University, 43210, USA
| | - Emily A Tomak
- School of Biology and Ecology, the University of Maine, 04469, USA
| | - Sadie L Waterman
- School of Biology and Ecology, the University of Maine, 04469, USA
| | - Kailee A Sprague
- School of Biology and Ecology, the University of Maine, 04469, USA
| | - Angelina White
- School of Biology and Ecology, the University of Maine, 04469, USA
| | | | - Sabrina M Varga
- School of Biology and Ecology, the University of Maine, 04469, USA
| | | | | | - Jared D Austin
- School of Biology and Ecology, the University of Maine, 04469, USA
| | | | - Troy E Hupper
- School of Biology and Ecology, the University of Maine, 04469, USA
| | - Joshua B Kelley
- Molecular and Biomedical Sciences, the University of Maine, 04469, USA
| | - Sharon L Amacher
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, 43210, USA
| | - Jared C Talbot
- School of Biology and Ecology, the University of Maine, 04469, USA
| |
Collapse
|
3
|
Danielli SG, Wei Y, Dyer MA, Stewart E, Sheppard H, Wachtel M, Schäfer BW, Patel AG, Langenau DM. Single cell transcriptomic profiling identifies tumor-acquired and therapy-resistant cell states in pediatric rhabdomyosarcoma. Nat Commun 2024; 15:6307. [PMID: 39060228 PMCID: PMC11282092 DOI: 10.1038/s41467-024-50527-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Rhabdomyosarcoma (RMS) is a pediatric tumor that resembles undifferentiated muscle cells; yet the extent to which cell state heterogeneity is shared with human development has not been described. Using single-cell/nucleus RNA sequencing from patient tumors, patient-derived xenografts, primary in vitro cultures, and cell lines, we identify four dominant muscle-lineage cell states: progenitor, proliferative, differentiated, and ground cells. We stratify these RMS cells/nuclei along the continuum of human muscle development and show that they share expression patterns with fetal/embryonal myogenic precursors rather than postnatal satellite cells. Fusion-negative RMS (FN-RMS) have a discrete stem cell hierarchy that recapitulates fetal muscle development and contain therapy-resistant FN-RMS progenitors that share transcriptomic similarity with bipotent skeletal mesenchymal cells. Fusion-positive RMS have tumor-acquired cells states, including a neuronal cell state, that are not found in myogenic development. This work identifies previously underappreciated cell state heterogeneity including unique treatment-resistant and tumor-acquired cell states that differ across RMS subtypes.
Collapse
Affiliation(s)
- Sara G Danielli
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, Zürich, Switzerland
| | - Yun Wei
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Elizabeth Stewart
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marco Wachtel
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, Zürich, Switzerland.
| | - Beat W Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, Zürich, Switzerland.
| | - Anand G Patel
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - David M Langenau
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
4
|
Robertson R, Li S, Filippelli RL, Chang NC. Muscle stem cell dysfunction in rhabdomyosarcoma and muscular dystrophy. Curr Top Dev Biol 2024; 158:83-121. [PMID: 38670717 DOI: 10.1016/bs.ctdb.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Muscle stem cells (MuSCs) are crucial to the repair and homeostasis of mature skeletal muscle. MuSC dysfunction and dysregulation of the myogenic program can contribute to the development of pathology ranging from cancers like rhabdomyosarcoma (RMS) or muscle degenerative diseases such as Duchenne muscular dystrophy (DMD). Both diseases exhibit dysregulation at nearly all steps of myogenesis. For instance, MuSC self-renewal processes are altered. In RMS, this leads to the creation of tumor propagating cells. In DMD, impaired asymmetric stem cell division creates a bias towards producing self-renewing stem cells instead of committing to differentiation. Hyperproliferation of these cells contribute to tumorigenesis in RMS and symmetric expansion of the self-renewing MuSC population in DMD. Both diseases also exhibit a repression of factors involved in terminal differentiation, halting RMS cells in the proliferative stage and thus driving tumor growth. Conversely, the MuSCs in DMD exhibit impaired differentiation and fuse prematurely, affecting myonuclei maturation and the integrity of the dystrophic muscle fiber. Finally, both disease states cause alterations to the MuSC niche. Various elements of the niche such as inflammatory and migratory signaling that impact MuSC behavior are dysregulated. Here we show how these seemingly distantly related diseases indeed have similarities in MuSC dysfunction, underlying the importance of considering MuSCs when studying the pathophysiology of muscle diseases.
Collapse
Affiliation(s)
- Rebecca Robertson
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Shulei Li
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Romina L Filippelli
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Natasha C Chang
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada.
| |
Collapse
|
5
|
Zarrabi A, Perrin D, Kavoosi M, Sommer M, Sezen S, Mehrbod P, Bhushan B, Machaj F, Rosik J, Kawalec P, Afifi S, Bolandi SM, Koleini P, Taheri M, Madrakian T, Łos MJ, Lindsey B, Cakir N, Zarepour A, Hushmandi K, Fallah A, Koc B, Khosravi A, Ahmadi M, Logue S, Orive G, Pecic S, Gordon JW, Ghavami S. Rhabdomyosarcoma: Current Therapy, Challenges, and Future Approaches to Treatment Strategies. Cancers (Basel) 2023; 15:5269. [PMID: 37958442 PMCID: PMC10650215 DOI: 10.3390/cancers15215269] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Rhabdomyosarcoma is a rare cancer arising in skeletal muscle that typically impacts children and young adults. It is a worldwide challenge in child health as treatment outcomes for metastatic and recurrent disease still pose a major concern for both basic and clinical scientists. The treatment strategies for rhabdomyosarcoma include multi-agent chemotherapies after surgical resection with or without ionization radiotherapy. In this comprehensive review, we first provide a detailed clinical understanding of rhabdomyosarcoma including its classification and subtypes, diagnosis, and treatment strategies. Later, we focus on chemotherapy strategies for this childhood sarcoma and discuss the impact of three mechanisms that are involved in the chemotherapy response including apoptosis, macro-autophagy, and the unfolded protein response. Finally, we discuss in vivo mouse and zebrafish models and in vitro three-dimensional bioengineering models of rhabdomyosarcoma to screen future therapeutic approaches and promote muscle regeneration.
Collapse
Affiliation(s)
- Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - David Perrin
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
| | - Mahboubeh Kavoosi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Micah Sommer
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
- Section of Physical Medicine and Rehabilitation, Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Serap Sezen
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Parvaneh Mehrbod
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Bhavya Bhushan
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Science, McGill University, Montreal, QC H3A 0C7, Canada
| | - Filip Machaj
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jakub Rosik
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Philip Kawalec
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Section of Neurosurgery, Department of Surgery, University of Manitoba, Health Sciences Centre, Winnipeg, MB R3A 1R9, Canada
| | - Saba Afifi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Seyed Mohammadreza Bolandi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Peiman Koleini
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Mohsen Taheri
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Benjamin Lindsey
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Nilufer Cakir
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran;
| | - Ali Fallah
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
| | - Bahattin Koc
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Türkiye
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Susan Logue
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01007 Vitoria-Gasteiz, Spain;
- University Institute for Regenerative Medicine and Oral Implantology–UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, USA;
| | - Joseph W. Gordon
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- College of Nursing, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555 Katowice, Poland
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
6
|
Ma X, Geng Z, Wang S, Yu Z, Liu T, Guan S, Du S, Zhu C. The driving mechanism and targeting value of mimicry between vascular endothelial cells and tumor cells in tumor progression. Biomed Pharmacother 2023; 165:115029. [PMID: 37343434 DOI: 10.1016/j.biopha.2023.115029] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023] Open
Abstract
The difficulty and poor prognosis of malignant tumor have always been a difficult problem to be solved. The internal components of solid tumor are complex, including tumor cells, stromal cells and immune cells, which play an important role in tumor proliferation, migration, metastasis and drug resistance. Hence, targeting of only the tumor cells will not likely improve survival. Various studies have reported that tumor cells and endothelial cells have high plasticity, which is reflected in the fact that they can simulate each other's characteristics by endothelial-mesenchymal transition (EndMT) and vasculogenic mimicry (VM). In this paper, this mutual mimicry concept was integrated and reviewed for the first time, and their similarities and implications for tumor development are discussed. At the same time, possible therapeutic methods are proposed to provide new directions and ideas for clinical targeted therapy and immunotherapy of tumor.
Collapse
Affiliation(s)
- Xiao Ma
- Department of Clinical Medicine, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, Liaoning 110001, China
| | - Ziang Geng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Sanhao Street 36, Heping District, Shenyang, Liaoning 110004, China
| | - Siqi Wang
- Department of Radiation Oncology, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, Liaoning 110001, China
| | - Zhongxue Yu
- Department of Cardiovascular Ultrasound, The First hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, Liaoning 110001, China
| | - Tiancong Liu
- Department of Otolaryngology, Shengjing Hospital of China Medical University, Sanhao Street 36, Heping District, Shenyang, Liaoning 110004, China.
| | - Shu Guan
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, Liaoning 110001, China.
| | - Shaonan Du
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Sanhao Street 36, Heping District, Shenyang, Liaoning 110004, China.
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, Liaoning 110001, China.
| |
Collapse
|
7
|
Chen J, Baxi K, Lipsitt AE, Hensch NR, Wang L, Sreenivas P, Modi P, Zhao XR, Baudin A, Robledo DG, Bandyopadhyay A, Sugalski A, Challa AK, Kurmashev D, Gilbert AR, Tomlinson GE, Houghton P, Chen Y, Hayes MN, Chen EY, Libich DS, Ignatius MS. Defining function of wild-type and three patient-specific TP53 mutations in a zebrafish model of embryonal rhabdomyosarcoma. eLife 2023; 12:e68221. [PMID: 37266578 PMCID: PMC10322150 DOI: 10.7554/elife.68221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/01/2023] [Indexed: 06/03/2023] Open
Abstract
In embryonal rhabdomyosarcoma (ERMS) and generally in sarcomas, the role of wild-type and loss- or gain-of-function TP53 mutations remains largely undefined. Eliminating mutant or restoring wild-type p53 is challenging; nevertheless, understanding p53 variant effects on tumorigenesis remains central to realizing better treatment outcomes. In ERMS, >70% of patients retain wild-type TP53, yet mutations when present are associated with worse prognosis. Employing a kRASG12D-driven ERMS tumor model and tp53 null (tp53-/-) zebrafish, we define wild-type and patient-specific TP53 mutant effects on tumorigenesis. We demonstrate that tp53 is a major suppressor of tumorigenesis, where tp53 loss expands tumor initiation from <35% to >97% of animals. Characterizing three patient-specific alleles reveals that TP53C176F partially retains wild-type p53 apoptotic activity that can be exploited, whereas TP53P153Δ and TP53Y220C encode two structurally related proteins with gain-of-function effects that predispose to head musculature ERMS. TP53P153Δ unexpectedly also predisposes to hedgehog-expressing medulloblastomas in the kRASG12D-driven ERMS-model.
Collapse
Affiliation(s)
- Jiangfei Chen
- Institute of Environmental Safety and Human Health, Wenzhou Medical UniversityWenzhouChina
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
| | - Kunal Baxi
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Amanda E Lipsitt
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Pediatrics, Division of Hematology Oncology, UT Health Sciences CenterSan AntonioUnited States
| | - Nicole Rae Hensch
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Long Wang
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Prethish Sreenivas
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Paulomi Modi
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Xiang Ru Zhao
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Antoine Baudin
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Biochemistry and Structural Biology, UT Health Sciences CenterSan AntonioUnited States
| | - Daniel G Robledo
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
| | - Abhik Bandyopadhyay
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
| | - Aaron Sugalski
- Department of Pediatrics, Division of Hematology Oncology, UT Health Sciences CenterSan AntonioUnited States
| | - Anil K Challa
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Biology, University of Alabama at BirminghamBirminghamUnited States
| | - Dias Kurmashev
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
| | - Andrea R Gilbert
- Department of Pathology and Laboratory Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Gail E Tomlinson
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Pediatrics, Division of Hematology Oncology, UT Health Sciences CenterSan AntonioUnited States
| | - Peter Houghton
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Yidong Chen
- Department of Population Health Sciences, UT Health Sciences CenterSan AntonioUnited States
| | - Madeline N Hayes
- Developmental and Stem Cell Biology, Hospital for Sick ChildrenTorontoCanada
| | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of WashingtonSeattleUnited States
| | - David S Libich
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Biochemistry and Structural Biology, UT Health Sciences CenterSan AntonioUnited States
| | - Myron S Ignatius
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| |
Collapse
|
8
|
Pomella S, Danielli SG, Alaggio R, Breunis WB, Hamed E, Selfe J, Wachtel M, Walters ZS, Schäfer BW, Rota R, Shipley JM, Hettmer S. Genomic and Epigenetic Changes Drive Aberrant Skeletal Muscle Differentiation in Rhabdomyosarcoma. Cancers (Basel) 2023; 15:2823. [PMID: 37345159 DOI: 10.3390/cancers15102823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma in children and adolescents, represents an aberrant form of skeletal muscle differentiation. Both skeletal muscle development, as well as regeneration of adult skeletal muscle are governed by members of the myogenic family of regulatory transcription factors (MRFs), which are deployed in a highly controlled, multi-step, bidirectional process. Many aspects of this complex process are deregulated in RMS and contribute to tumorigenesis. Interconnected loops of super-enhancers, called core regulatory circuitries (CRCs), define aberrant muscle differentiation in RMS cells. The transcriptional regulation of MRF expression/activity takes a central role in the CRCs active in skeletal muscle and RMS. In PAX3::FOXO1 fusion-positive (PF+) RMS, CRCs maintain expression of the disease-driving fusion oncogene. Recent single-cell studies have revealed hierarchically organized subsets of cells within the RMS cell pool, which recapitulate developmental myogenesis and appear to drive malignancy. There is a large interest in exploiting the causes of aberrant muscle development in RMS to allow for terminal differentiation as a therapeutic strategy, for example, by interrupting MEK/ERK signaling or by interfering with the epigenetic machinery controlling CRCs. In this review, we provide an overview of the genetic and epigenetic framework of abnormal muscle differentiation in RMS, as it provides insights into fundamental mechanisms of RMS malignancy, its remarkable phenotypic diversity and, ultimately, opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Silvia Pomella
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS Istituto Ospedale Pediatrico Bambino Gesu, Viale San Paolo 15, 00146 Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Sara G Danielli
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, 8032 Zürich, Switzerland
| | - Rita Alaggio
- Department of Pathology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy
| | - Willemijn B Breunis
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, 8032 Zürich, Switzerland
| | - Ebrahem Hamed
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, 79106 Freiburg, Germany
| | - Joanna Selfe
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London SM2 FNG, UK
| | - Marco Wachtel
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, 8032 Zürich, Switzerland
| | - Zoe S Walters
- Translational Epigenomics Team, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Beat W Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, 8032 Zürich, Switzerland
| | - Rossella Rota
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS Istituto Ospedale Pediatrico Bambino Gesu, Viale San Paolo 15, 00146 Rome, Italy
| | - Janet M Shipley
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London SM2 FNG, UK
| | - Simone Hettmer
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), 79104 Freiburg, Germany
- Comprehensive Cancer Centre Freiburg (CCCF), University Medical Center Freiburg, 790106 Freiburg, Germany
| |
Collapse
|
9
|
Fritzke M, Chen K, Tang W, Stinson S, Pham T, Wang Y, Xu L, Chen EY. The MYC-YBX1 Circuit in Maintaining Stem-like Vincristine-Resistant Cells in Rhabdomyosarcoma. Cancers (Basel) 2023; 15:2788. [PMID: 37345125 DOI: 10.3390/cancers15102788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a pediatric soft tissue sarcoma that causes significant devastation, with no effective therapy for relapsed disease. The mechanisms behind treatment failures are poorly understood. Our study showed that treatment of RMS cells with vincristine led to an increase in CD133-positive stem-like resistant cells. Single cell RNAseq analysis revealed that MYC and YBX1 were among the top-scoring transcription factors in CD133-high expressing cells. Targeting MYC and YBX1 using CRISPR/Cas9 reduced stem-like characteristics and viability of the vincristine-resistant cells. MYC and YBX1 showed mutual regulation, with MYC binding to the YBX1 promoter and YBX1 binding to MYC mRNA. The MYC inhibitor MYC361i synergized with vincristine to reduce tumor growth and stem-like cells in a zebrafish model of RMS. MYC and YBX expression showed a positive correlation in RMS patients, and high MYC expression correlated with poor survival. Targeting the MYC-YBX1 axis holds promise for improving survival in RMS patients.
Collapse
Affiliation(s)
- Madeline Fritzke
- Department of Laboratory Pathology and Medicine, University of Washington, Seattle, WA 98195, USA
| | - Kenian Chen
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Weiliang Tang
- Department of Laboratory Pathology and Medicine, University of Washington, Seattle, WA 98195, USA
| | - Spencer Stinson
- Department of Laboratory Pathology and Medicine, University of Washington, Seattle, WA 98195, USA
| | - Thao Pham
- Department of Laboratory Pathology and Medicine, University of Washington, Seattle, WA 98195, USA
- Astellas US Technologies, Universal Cells, Inc., Seattle, WA 98121, USA
| | - Yadong Wang
- Department of Laboratory Pathology and Medicine, University of Washington, Seattle, WA 98195, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eleanor Y Chen
- Department of Laboratory Pathology and Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
10
|
Proietto M, Crippa M, Damiani C, Pasquale V, Sacco E, Vanoni M, Gilardi M. Tumor heterogeneity: preclinical models, emerging technologies, and future applications. Front Oncol 2023; 13:1164535. [PMID: 37188201 PMCID: PMC10175698 DOI: 10.3389/fonc.2023.1164535] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Heterogeneity describes the differences among cancer cells within and between tumors. It refers to cancer cells describing variations in morphology, transcriptional profiles, metabolism, and metastatic potential. More recently, the field has included the characterization of the tumor immune microenvironment and the depiction of the dynamics underlying the cellular interactions promoting the tumor ecosystem evolution. Heterogeneity has been found in most tumors representing one of the most challenging behaviors in cancer ecosystems. As one of the critical factors impairing the long-term efficacy of solid tumor therapy, heterogeneity leads to tumor resistance, more aggressive metastasizing, and recurrence. We review the role of the main models and the emerging single-cell and spatial genomic technologies in our understanding of tumor heterogeneity, its contribution to lethal cancer outcomes, and the physiological challenges to consider in designing cancer therapies. We highlight how tumor cells dynamically evolve because of the interactions within the tumor immune microenvironment and how to leverage this to unleash immune recognition through immunotherapy. A multidisciplinary approach grounded in novel bioinformatic and computational tools will allow reaching the integrated, multilayered knowledge of tumor heterogeneity required to implement personalized, more efficient therapies urgently required for cancer patients.
Collapse
Affiliation(s)
- Marco Proietto
- Next Generation Sequencing Core, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Martina Crippa
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Imaging Center, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Chiara Damiani
- Infrastructure Systems Biology Europe /Centre of Systems Biology (ISBE/SYSBIO) Centre of Systems Biology, Milan, Italy
- Department of Biotechnology and Biosciences, School of Sciences, University of Milano-Bicocca, Milan, Italy
| | - Valentina Pasquale
- Infrastructure Systems Biology Europe /Centre of Systems Biology (ISBE/SYSBIO) Centre of Systems Biology, Milan, Italy
- Department of Biotechnology and Biosciences, School of Sciences, University of Milano-Bicocca, Milan, Italy
| | - Elena Sacco
- Infrastructure Systems Biology Europe /Centre of Systems Biology (ISBE/SYSBIO) Centre of Systems Biology, Milan, Italy
- Department of Biotechnology and Biosciences, School of Sciences, University of Milano-Bicocca, Milan, Italy
| | - Marco Vanoni
- Infrastructure Systems Biology Europe /Centre of Systems Biology (ISBE/SYSBIO) Centre of Systems Biology, Milan, Italy
- Department of Biotechnology and Biosciences, School of Sciences, University of Milano-Bicocca, Milan, Italy
| | - Mara Gilardi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Salk Cancer Center, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
11
|
Wei Y, Qin Q, Yan C, Hayes MN, Garcia SP, Xi H, Do D, Jin AH, Eng TC, McCarthy KM, Adhikari A, Onozato ML, Spentzos D, Neilsen GP, Iafrate AJ, Wexler LH, Pyle AD, Suvà ML, Dela Cruz F, Pinello L, Langenau DM. Single-cell analysis and functional characterization uncover the stem cell hierarchies and developmental origins of rhabdomyosarcoma. NATURE CANCER 2022; 3:961-975. [PMID: 35982179 PMCID: PMC10430812 DOI: 10.1038/s43018-022-00414-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 06/24/2022] [Indexed: 04/29/2023]
Abstract
Rhabdomyosarcoma (RMS) is a common childhood cancer that shares features with developing skeletal muscle. Yet, the conservation of cellular hierarchy with human muscle development and the identification of molecularly defined tumor-propagating cells has not been reported. Using single-cell RNA-sequencing, DNA-barcode cell fate mapping and functional stem cell assays, we uncovered shared tumor cell hierarchies in RMS and human muscle development. We also identified common developmental stages at which tumor cells become arrested. Fusion-negative RMS cells resemble early myogenic cells found in embryonic and fetal development, while fusion-positive RMS cells express a highly specific gene program found in muscle cells transiting from embryonic to fetal development at 7-7.75 weeks of age. Fusion-positive RMS cells also have neural pathway-enriched states, suggesting less-rigid adherence to muscle-lineage hierarchies. Finally, we identified a molecularly defined tumor-propagating subpopulation in fusion-negative RMS that shares remarkable similarity to bi-potent, muscle mesenchyme progenitors that can make both muscle and osteogenic cells.
Collapse
Affiliation(s)
- Yun Wei
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Qian Qin
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Chuan Yan
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Madeline N Hayes
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Sara P Garcia
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
| | - Haibin Xi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
| | - Daniel Do
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Alexander H Jin
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Tiffany C Eng
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Karin M McCarthy
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Abhinav Adhikari
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Maristela L Onozato
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Dimitrios Spentzos
- Center for Sarcoma and Connective Tissue Oncology, Department of Orthopedic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Gunnlaugur P Neilsen
- Center for Sarcoma and Connective Tissue Oncology, Department of Orthopedic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - A John Iafrate
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Leonard H Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - April D Pyle
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
| | - Mario L Suvà
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Filemon Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luca Pinello
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA.
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA.
| | - David M Langenau
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA.
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA.
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
12
|
Kwiatkowska I, Hermanowicz JM, Iwinska Z, Kowalczuk K, Iwanowska J, Pawlak D. Zebrafish—An Optimal Model in Experimental Oncology. Molecules 2022; 27:molecules27134223. [PMID: 35807468 PMCID: PMC9268704 DOI: 10.3390/molecules27134223] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/10/2022] [Accepted: 06/28/2022] [Indexed: 02/02/2023] Open
Abstract
A thorough understanding of cancer pathogenesis is a necessary step in the development of more effective and safer therapy. However, due to the complexity of the process and intricate interactions, studying tumor development is an extremely difficult and challenging task. In bringing this issue closer, different scientific models with various advancement levels are helpful. Cell cultures is a system that is too simple and does not allow for multidirectional research. On the other hand, rodent models, although commonly used, are burdened with several limitations. For this reason, new model organisms that will allow for the studying of carcinogenesis stages and factors reliably involved in them are urgently sought after. Danio rerio, an inconspicuous fish endowed with unique features, is gaining in importance in the world of scientific research. Including it in oncological research brings solutions to many challenges afflicting modern medicine. This article aims to illustrate the usefulness of Danio rerio as a model organism which turns out to be a powerful and unique tool for studying the stages of carcinogenesis and solving the hitherto incomprehensible processes that lead to the development of the disease.
Collapse
Affiliation(s)
- Iwona Kwiatkowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
- Correspondence: ; Tel./Fax: +48-8574-856-01
| | - Justyna Magdalena Hermanowicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
- Department of Clinical Pharmacy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland
| | - Zaneta Iwinska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| | - Krystyna Kowalczuk
- Department of Integrated Medical Care, Medical University of Bialystok, ul. M Skłodowskiej-Curie 7A, 15-096 Bialystok, Poland;
| | - Jolanta Iwanowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| |
Collapse
|
13
|
Patel AG, Chen X, Huang X, Clay MR, Komorova N, Krasin MJ, Pappo A, Tillman H, Orr BA, McEvoy J, Gordon B, Blankenship K, Reilly C, Zhou X, Norrie JL, Karlstrom A, Yu J, Wodarz D, Stewart E, Dyer MA. The myogenesis program drives clonal selection and drug resistance in rhabdomyosarcoma. Dev Cell 2022; 57:1226-1240.e8. [PMID: 35483358 PMCID: PMC9133224 DOI: 10.1016/j.devcel.2022.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 02/07/2022] [Accepted: 04/01/2022] [Indexed: 11/29/2022]
Abstract
Rhabdomyosarcoma (RMS) is a pediatric cancer with features of skeletal muscle; patients with unresectable or metastatic RMS fare poorly due to high rates of disease recurrence. Here, we use single-cell and single-nucleus RNA sequencing to show that RMS tumors recapitulate the spectrum of embryonal myogenesis. Using matched patient samples from a clinical trial and orthotopic patient-derived xenografts (O-PDXs), we show that chemotherapy eliminates the most proliferative component with features of myoblasts within embryonal RMS; after treatment, the immature population with features of paraxial mesoderm expands to reconstitute the developmental hierarchy of the original tumor. We discovered that this paraxial mesoderm population is dependent on EGFR signaling and is sensitive to EGFR inhibitors. Taken together, these data serve as a proof of concept that targeting each developmental state in embryonal RMS is an effective strategy for improving outcomes by preventing disease recurrence.
Collapse
Affiliation(s)
- Anand G Patel
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xin Huang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael R Clay
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Natalia Komorova
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Matthew J Krasin
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alberto Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather Tillman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Justina McEvoy
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brittney Gordon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kaley Blankenship
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Colleen Reilly
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jackie L Norrie
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Asa Karlstrom
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Dominik Wodarz
- Department of Population Health and Disease Prevention, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA
| | - Elizabeth Stewart
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
14
|
Context-dependent modulation of aggressiveness of pediatric tumors by individual oncogenic RAS isoforms. Oncogene 2021; 40:4955-4966. [PMID: 34172934 PMCID: PMC8342309 DOI: 10.1038/s41388-021-01904-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/25/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023]
Abstract
A prototypic pediatric cancer that frequently shows activation of RAS signaling is embryonal rhabdomyosarcoma (ERMS). ERMS also show aberrant Hedgehog (HH)/GLI signaling activity and can be driven by germline mutations in this pathway. We show, that in ERMS cell lines derived from sporadic tumors i.e. from tumors not caused by an inherited genetic variant, HH/GLI signaling plays a subordinate role, because oncogenic mutations in HRAS, KRAS, or NRAS (collectively named oncRAS) inhibit the main HH target GLI1 via the MEK/ERK-axis, but simultaneously increase proliferation and tumorigenicity. oncRAS also modulate expression of stem cell markers in an isoform- and context-dependent manner. In Hh-driven murine ERMS that are caused by a Patched mutation, oncHRAS and mainly oncKRAS accelerate tumor development, whereas oncNRAS induces a more differentiated phenotype. These features occur when the oncRAS mutations are induced at the ERMS precursor stage, but not when induced in already established tumors. Moreover, in contrast to what is seen in human cell lines, oncRAS mutations do not alter Hh signaling activity and marginally affect expression of stem cell markers. Together, all three oncRAS mutations seem to be advantageous for ERMS cell lines despite inhibition of HH signaling and isoform-specific modulation of stem cell markers. In contrast, oncRAS mutations do not inhibit Hh-signaling in Hh-driven ERMS. In this model, oncRAS mutations seem to be advantageous for specific ERMS populations that occur within a specific time window during ERMS development. In addition, this window may be different for individual oncRAS isoforms, at least in the mouse.
Collapse
|
15
|
Lipsitt A, Hensch NR, Moreno-Campos R, Baxi K, Chen J, Challa AK, Chen EY, Ignatius MS. Zebrafish Tumor Graft Transplantation to Grow Tumors In Vivo That Engraft Poorly as Single Cell Suspensions. Zebrafish 2021; 18:293-296. [PMID: 34030492 DOI: 10.1089/zeb.2021.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Angiosarcoma is a clinically aggressive tumor with a high rate of mortality. It can arise in vascular or lymphatic tissues, involve any part of the body, and aggressively spread locally or metastasize. Angiosarcomas spontaneously develop in the tp53 deleted (tp53del/del) zebrafish mutant. However, established protocols for tumor dissection and transplantation of single cell suspensions of angiosarcoma tumors result in inferior implantation rates. To resolve these complications, we developed a new tumor grafting technique for engraftment of angiosarcoma and similar tumors in zebrafish, which maintains the tumor microenvironment and has superior rates of engraftment.
Collapse
Affiliation(s)
- Amanda Lipsitt
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
- Department of Pediatrics, Division of Hematology Oncology, UT Health Sciences Center, San Antonio, Texas, USA
| | - Nicole R Hensch
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
| | - Rodrigo Moreno-Campos
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
| | - Kunal Baxi
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
| | - Jiangfei Chen
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
- Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, P.R. China
| | - Anil K Challa
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Myron S Ignatius
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
| |
Collapse
|
16
|
Her ZP, Yeo KS, Howe C, Levee T, Zhu S. Zebrafish Model of Neuroblastoma Metastasis. J Vis Exp 2021. [PMID: 33779609 DOI: 10.3791/62416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Zebrafish has emerged as an important animal model to study human diseases, especially cancer. Along with the robust transgenic and genome editing technologies applied in zebrafish modeling, the ease of maintenance, high-yield productivity, and powerful live imaging altogether make the zebrafish a valuable model system to study metastasis and cellular and molecular bases underlying this process in vivo. The first zebrafish neuroblastoma (NB) model of metastasis was developed by overexpressing two oncogenes, MYCN and LMO1, under control of the dopamine-beta-hydroxylase (dβh) promoter. Co-overexpressed MYCN and LMO1 led to the reduced latency and increased penetrance of neuroblastomagenesis, as well as accelerated distant metastasis of tumor cells. This new model reliably reiterates many key features of human metastatic NB, including involvement of clinically relevant and metastasis-associated genetic alterations; natural and spontaneous development of metastasis in vivo; and conserved sites of metastases. Therefore, the zebrafish model possesses unique advantages to dissect the complex process of tumor metastasis in vivo.
Collapse
Affiliation(s)
- Zuag Paj Her
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center
| | - Kok Siong Yeo
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center
| | - Cassie Howe
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center
| | - Taylor Levee
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center;
| |
Collapse
|
17
|
Abstract
Zebrafish are rapidly becoming a leading model organism for cancer research. The genetic pathways driving cancer are highly conserved between zebrafish and humans, and the ability to easily manipulate the zebrafish genome to rapidly generate transgenic animals makes zebrafish an excellent model organism. Transgenic zebrafish containing complex, patient-relevant genotypes have been used to model many cancer types. Here we present a comprehensive review of transgenic zebrafish cancer models as a resource to the field and highlight important areas of cancer biology that have yet to be studied in the fish. The ability to image cancer cells and niche biology in an endogenous tumor makes zebrafish an indispensable model organism in which we can further understand the mechanisms that drive tumorigenesis and screen for potential new cancer therapies.
Collapse
Affiliation(s)
- Alicia M. McConnell
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Haley R. Noonan
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Leonard I. Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02138, USA
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Stem Cell and Regenerative Biology Department and Howard Hughes Medical Institute, Harvard University, Boston, Massachusetts 02138, USA
| |
Collapse
|
18
|
Chen X, Li Y, Yao T, Jia R. Benefits of Zebrafish Xenograft Models in Cancer Research. Front Cell Dev Biol 2021; 9:616551. [PMID: 33644052 PMCID: PMC7905065 DOI: 10.3389/fcell.2021.616551] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
As a promising in vivo tool for cancer research, zebrafish have been widely applied in various tumor studies. The zebrafish xenograft model is a low-cost, high-throughput tool for cancer research that can be established quickly and requires only a small sample size, which makes it favorite among researchers. Zebrafish patient-derived xenograft (zPDX) models provide promising evidence for short-term clinical treatment. In this review, we discuss the characteristics and advantages of zebrafish, such as their transparent and translucent features, the use of vascular fluorescence imaging, the establishment of metastatic and intracranial orthotopic models, individual pharmacokinetics measurements, and tumor microenvironment. Furthermore, we introduce how these characteristics and advantages are applied other in tumor studies. Finally, we discuss the future direction of the use of zebrafish in tumor studies and provide new ideas for the application of it.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Tengteng Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
19
|
Abstract
Tumor models allowing for the in vivo investigation of molecular mechanisms driving tumor progression and metastasis are important to develop novel strategies for cancer treatment. Unfortunately, for Ewing sarcoma no adequate genetic animal models are currently available. Mouse xenograft models are the state of the art to model Ewing sarcoma in vivo. Here, we describe an alternative Ewing sarcoma xenograft model in embryonic and larval zebrafish. This xenograft model offers live imaging and easy compound testing opportunities hereby complementing mouse xenograft models. In this chapter, we provide a detailed protocol how to xenograft Ewing sarcoma cells (shSK-E17T) into 2-day-old zebrafish and how xenografted zebrafish can be imaged and analyzed over consecutive days to study tumor proliferation.
Collapse
|
20
|
HDAC6 promotes growth, migration/invasion, and self-renewal of rhabdomyosarcoma. Oncogene 2020; 40:578-591. [PMID: 33199827 PMCID: PMC7855743 DOI: 10.1038/s41388-020-01550-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 10/21/2020] [Accepted: 10/30/2020] [Indexed: 01/20/2023]
Abstract
Rhabdomyosarcoma (RMS) is a devastating pediatric sarcoma. The survival outcomes remain poor for patients with relapsed or metastatic disease. Effective targeted therapy is lacking due to our limited knowledge of the underlying cellular and molecular mechanisms leading to disease progression. In this study, we used functional assays in vitro and in vivo (zebrafish and xenograft mouse models) to demonstrate the crucial role of HDAC6, a cytoplasmic histone deacetylase, in driving RMS tumor growth, self-renewal, and migration/invasion. Treatment with HDAC6-selective inhibitors recapitulates the HDAC6 loss-of-function phenotypes. HDAC6 regulates cytoskeletal dynamics to promote tumor cell migration and invasion. RAC1, a Rho family GTPase, is an essential mediator of HDAC6 function, and is necessary and sufficient for RMS cell migration and invasion. High expression of RAC1 correlates with poor clinical prognosis in RMS patients. Targeting the HDAC6-RAC1 axis represents a promising therapeutic option for improving survival outcomes of RMS patients.
Collapse
|
21
|
Abstract
Rhabdomyosarcoma (RMS) is an aggressive childhood mesenchymal tumor with two major molecular and histopathologic subtypes: fusion-positive (FP)RMS, characterized by the PAX3-FOXO1 fusion protein and largely of alveolar histology, and fusion-negative (FN)RMS, the majority of which exhibit embryonal tumor histology. Metastatic disease continues to be associated with poor overall survival despite intensive treatment strategies. Studies on RMS biology have provided some insight into autocrine as well as paracrine signaling pathways that contribute to invasion and metastatic propensity. Such pathways include those driven by the PAX3-FOXO1 fusion oncoprotein in FPRMS and signaling pathways such as IGF/RAS/MEK/ERK, PI3K/AKT/mTOR, cMET, FGFR4, and PDGFR in both FP and FNRMS. In addition, specific cytoskeletal proteins, G protein coupled receptors, Hedgehog, Notch, Wnt, Hippo, and p53 pathways play a role, as do specific microRNA. Paracrine factors, including secreted proteins and RMS-derived exosomes that carry cargo of protein and miRNA, have also recently emerged as potentially important players in RMS biology. This review summarizes the known factors contributing to RMS invasion and metastasis and their implications on identifying targets for treatment and a better understanding of metastatic RMS.
Collapse
|
22
|
Manzella G, Schreck LD, Breunis WB, Molenaar J, Merks H, Barr FG, Sun W, Römmele M, Zhang L, Tchinda J, Ngo QA, Bode P, Delattre O, Surdez D, Rekhi B, Niggli FK, Schäfer BW, Wachtel M. Phenotypic profiling with a living biobank of primary rhabdomyosarcoma unravels disease heterogeneity and AKT sensitivity. Nat Commun 2020; 11:4629. [PMID: 32934208 PMCID: PMC7492191 DOI: 10.1038/s41467-020-18388-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer therapy is currently shifting from broadly used cytotoxic drugs to patient-specific precision therapies. Druggable driver oncogenes, identified by molecular analyses, are present in only a subset of patients. Functional profiling of primary tumor cells could circumvent these limitations, but suitable platforms are unavailable for most cancer entities. Here, we describe an in vitro drug profiling platform for rhabdomyosarcoma (RMS), using a living biobank composed of twenty RMS patient-derived xenografts (PDX) for high-throughput drug testing. Optimized in vitro conditions preserve phenotypic and molecular characteristics of primary PDX cells and are compatible with propagation of cells directly isolated from patient tumors. Besides a heterogeneous spectrum of responses of largely patient-specific vulnerabilities, profiling with a large drug library reveals a strong sensitivity towards AKT inhibitors in a subgroup of RMS. Overall, our study highlights the feasibility of in vitro drug profiling of primary RMS for patient-specific treatment selection in a co-clinical setting. Patient-specific precision medicine approaches are important for future cancer therapies. Here, the authors show that functional drug profiling with Rhabdomyosarcoma cells isolated from PDX and primary patient tumors uncovers patient-specific vulnerabilities.
Collapse
Affiliation(s)
- Gabriele Manzella
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | - Leonie D Schreck
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | - Willemijn B Breunis
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland.,Princess Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584, CT, Utrecht, The Netherlands
| | - Jan Molenaar
- Princess Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584, CT, Utrecht, The Netherlands
| | - Hans Merks
- Princess Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584, CT, Utrecht, The Netherlands
| | - Frederic G Barr
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Wenyue Sun
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Michaela Römmele
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | - Luduo Zhang
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | - Joelle Tchinda
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | - Quy A Ngo
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | - Peter Bode
- University Hospital Zurich, Institute of Surgical Pathology, Schmelzbergstrasse 12, CH-8091, Zurich, Switzerland
| | - Olivier Delattre
- France INSERM U830, Équipe Labellisé LNCC, PSL Université, SIREDO Oncology Centre, Institut Curie, Paris, France
| | - Didier Surdez
- France INSERM U830, Équipe Labellisé LNCC, PSL Université, SIREDO Oncology Centre, Institut Curie, Paris, France
| | - Bharat Rekhi
- Tata Memorial Hospital, Department of Pathology, Dr E.B. road, Parel, Mumbai, 400012, India
| | - Felix K Niggli
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | - Beat W Schäfer
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland.
| | - Marco Wachtel
- University Children's Hospital, Department of Oncology and Children's Research Center, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| |
Collapse
|
23
|
Chong JX, Talbot JC, Teets EM, Previs S, Martin BL, Shively KM, Marvin CT, Aylsworth AS, Saadeh-Haddad R, Schatz UA, Inzana F, Ben-Omran T, Almusafri F, Al-Mulla M, Buckingham KJ, Harel T, Mor-Shaked H, Radhakrishnan P, Girisha KM, Nayak SS, Shukla A, Dieterich K, Faure J, Rendu J, Capri Y, Latypova X, Nickerson DA, Warshaw DM, Janssen PM, Amacher SL, Bamshad MJ, Bamshad MJ. Mutations in MYLPF Cause a Novel Segmental Amyoplasia that Manifests as Distal Arthrogryposis. Am J Hum Genet 2020; 107:293-310. [PMID: 32707087 DOI: 10.1016/j.ajhg.2020.06.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
We identified ten persons in six consanguineous families with distal arthrogryposis (DA) who had congenital contractures, scoliosis, and short stature. Exome sequencing revealed that each affected person was homozygous for one of two different rare variants (c.470G>T [p.Cys157Phe] or c.469T>C [p.Cys157Arg]) affecting the same residue of myosin light chain, phosphorylatable, fast skeletal muscle (MYLPF). In a seventh family, a c.487G>A (p.Gly163Ser) variant in MYLPF arose de novo in a father, who transmitted it to his son. In an eighth family comprised of seven individuals with dominantly inherited DA, a c.98C>T (p.Ala33Val) variant segregated in all four persons tested. Variants in MYLPF underlie both dominant and recessively inherited DA. Mylpf protein models suggest that the residues associated with dominant DA interact with myosin whereas the residues altered in families with recessive DA only indirectly impair this interaction. Pathological and histological exam of a foot amputated from an affected child revealed complete absence of skeletal muscle (i.e., segmental amyoplasia). To investigate the mechanism for this finding, we generated an animal model for partial MYLPF impairment by knocking out zebrafish mylpfa. The mylpfa mutant had reduced trunk contractile force and complete pectoral fin paralysis, demonstrating that mylpf impairment most severely affects limb movement. mylpfa mutant muscle weakness was most pronounced in an appendicular muscle and was explained by reduced myosin activity and fiber degeneration. Collectively, our findings demonstrate that partial loss of MYLPF function can lead to congenital contractures, likely as a result of degeneration of skeletal muscle in the distal limb.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Michael J Bamshad
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Seattle Children's Hospital, Seattle, WA 98105, USA.
| |
Collapse
|
24
|
Raby L, Völkel P, Le Bourhis X, Angrand PO. Genetic Engineering of Zebrafish in Cancer Research. Cancers (Basel) 2020; 12:E2168. [PMID: 32759814 PMCID: PMC7464884 DOI: 10.3390/cancers12082168] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022] Open
Abstract
Zebrafish (Danio rerio) is an excellent model to study a wide diversity of human cancers. In this review, we provide an overview of the genetic and reverse genetic toolbox allowing the generation of zebrafish lines that develop tumors. The large spectrum of genetic tools enables the engineering of zebrafish lines harboring precise genetic alterations found in human patients, the generation of zebrafish carrying somatic or germline inheritable mutations or zebrafish showing conditional expression of the oncogenic mutations. Comparative transcriptomics demonstrate that many of the zebrafish tumors share molecular signatures similar to those found in human cancers. Thus, zebrafish cancer models provide a unique in vivo platform to investigate cancer initiation and progression at the molecular and cellular levels, to identify novel genes involved in tumorigenesis as well as to contemplate new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Pierre-Olivier Angrand
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (L.R.); (P.V.); (X.L.B.)
| |
Collapse
|
25
|
Abstract
Metastasis, the dispersal of cancer cells from a primary tumor to secondary sites within the body, is the leading cause of cancer-related death. Animal models have been an indispensable tool to investigate the complex interactions between the cancer cells and the tumor microenvironment during the metastatic cascade. The zebrafish (Danio rerio) has emerged as a powerful vertebrate model for studying metastatic events in vivo. The zebrafish has many attributes including ex-utero development, which facilitates embryonic manipulation, as well as optically transparent tissues, which enables in vivo imaging of fluorescently labeled cells in real time. Here, we summarize the techniques which have been used to study cancer biology and metastasis in the zebrafish model organism, including genetic manipulation and transgenesis, cell transplantation, live imaging, and high-throughput compound screening. Finally, we discuss studies using the zebrafish, which have complemented and benefited metastasis research.
Collapse
Affiliation(s)
- Katy R Astell
- The Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Dirk Sieger
- The Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| |
Collapse
|
26
|
Hromowyk KJ, Talbot JC, Martin BL, Janssen PML, Amacher SL. Cell fusion is differentially regulated in zebrafish post-embryonic slow and fast muscle. Dev Biol 2020; 462:85-100. [PMID: 32165147 PMCID: PMC7225055 DOI: 10.1016/j.ydbio.2020.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 02/08/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Skeletal muscle fusion occurs during development, growth, and regeneration. To investigate how muscle fusion compares among different muscle cell types and developmental stages, we studied muscle cell fusion over time in wild-type, myomaker (mymk), and jam2a mutant zebrafish. Using live imaging, we show that embryonic myoblast elongation and fusion correlate tightly with slow muscle cell migration. In wild-type embryos, only fast muscle fibers are multinucleate, consistent with previous work showing that the cell fusion regulator gene mymk is specifically expressed throughout the embryonic fast muscle domain. However, by 3 weeks post-fertilization, slow muscle fibers also become multinucleate. At this late-larval stage, mymk is not expressed in muscle fibers, but is expressed in small cells near muscle fibers. Although previous work showed that both mymk and jam2a are required for embryonic fast muscle cell fusion, we observe that muscle force and function is almost normal in mymk and jam2a mutant embryos, despite the lack of fast muscle multinucleation. We show that genetic requirements change post-embryonically, with jam2a becoming much less important by late-larval stages and mymk now required for muscle fusion and growth in both fast and slow muscle cell types. Correspondingly, adult mymk mutants perform poorly in sprint and endurance tests compared to wild-type and jam2a mutants. We show that adult mymk mutant muscle contains small mononucleate myofibers with average myonuclear domain size equivalent to that in wild type adults. The mymk mutant fibers have decreased Laminin expression and increased numbers of Pax7-positive cells, suggesting that impaired fiber growth and active regeneration contribute to the muscle phenotype. Our findings identify several aspects of muscle fusion that change with time in slow and fast fibers as zebrafish develop beyond embryonic stages.
Collapse
Affiliation(s)
- Kimberly J Hromowyk
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Jared C Talbot
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA.
| | - Brit L Martin
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Paul M L Janssen
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Sharon L Amacher
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Center for RNA Biology, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
27
|
Pham T, Robinson K, Vleeshouwer-Neumann T, Annis JE, Chen EY. Characterization of GRK5 as a novel regulator of rhabdomyosarcoma tumor cell growth and self-renewal. Oncotarget 2020; 11:1448-1461. [PMID: 32363002 PMCID: PMC7185065 DOI: 10.18632/oncotarget.27562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/03/2020] [Indexed: 12/24/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft-tissue pediatric sarcoma. Clinical outcomes for RMS patients with relapsed or metastatic disease remain poor. Treatment options remain limited, presenting an urgent need for novel therapeutic targets. Using a high-throughput siRNA screen against the human kinome, we identified GRK5, a G-protein receptor kinase, as a novel regulator of RMS tumor cell growth and self-renewal. Through functional assays in vitro and in vivo, we show that GRK5 regulates cell cycle in a kinase-independent manner to promote RMS tumor cell growth. NFAT1 expression is regulated by GRK5 in a kinase independent manner, and loss of NFAT1 phenocopies GRK5 loss-of-function effects on the cell cycle alterations. Self-renewal of tumor propagating cells (TPCs) is thought to give rise to tumor relapse. We show that loss of GRK5 results in a significant reduction of RMS self-renewal capacity in part due to increased cell death. Treatment of human RMS xenografts in mice with CCG-215022, a GRK5-selective inhibitor, results in reduced tumor growth and self-renewal in both major subtypes of RMS. GRK5 represents a novel therapeutic target for the treatment of RMS.
Collapse
Affiliation(s)
- Thao Pham
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Kristin Robinson
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - James E. Annis
- Quellos HTS Core, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Eleanor Y. Chen
- Department of Pathology, University of Washington, Seattle, WA, USA
| |
Collapse
|
28
|
Elliot A, Myllymäki H, Feng Y. Inflammatory Responses during Tumour Initiation: From Zebrafish Transgenic Models of Cancer to Evidence from Mouse and Man. Cells 2020; 9:cells9041018. [PMID: 32325966 PMCID: PMC7226149 DOI: 10.3390/cells9041018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The zebrafish is now an important model organism for cancer biology studies and provides unique and complementary opportunities in comparison to the mammalian equivalent. The translucency of zebrafish has allowed in vivo live imaging studies of tumour initiation and progression at the cellular level, providing novel insights into our understanding of cancer. Here we summarise the available transgenic zebrafish tumour models and discuss what we have gleaned from them with respect to cancer inflammation. In particular, we focus on the host inflammatory response towards transformed cells during the pre-neoplastic stage of tumour development. We discuss features of tumour-associated macrophages and neutrophils in mammalian models and present evidence that supports the idea that these inflammatory cells promote early stage tumour development and progression. Direct live imaging of tumour initiation in zebrafish models has shown that the intrinsic inflammation induced by pre-neoplastic cells is tumour promoting. Signals mediating leukocyte recruitment to pre-neoplastic cells in zebrafish correspond to the signals that mediate leukocyte recruitment in mammalian tumours. The activation state of macrophages and neutrophils recruited to pre-neoplastic cells in zebrafish appears to be heterogenous, as seen in mammalian models, which provides an opportunity to study the plasticity of innate immune cells during tumour initiation. Although several potential mechanisms are described that might mediate the trophic function of innate immune cells during tumour initiation in zebrafish, there are several unknowns that are yet to be resolved. Rapid advancement of genetic tools and imaging technologies for zebrafish will facilitate research into the mechanisms that modulate leukocyte function during tumour initiation and identify targets for cancer prevention.
Collapse
Affiliation(s)
| | | | - Yi Feng
- Correspondence: ; Tel.: +44-(0)131-242-6685
| |
Collapse
|
29
|
Dawson LE, D'Agostino L, Hakim AA, Lackman RD, Brown SA, Sensenig RB, Antonello ZA, Kuzin II. Induction of Myogenic Differentiation Improves Chemosensitivity of Chemoresistant Cells in Soft-Tissue Sarcoma Cell Lines. Sarcoma 2020; 2020:8647981. [PMID: 32300280 PMCID: PMC7136814 DOI: 10.1155/2020/8647981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/30/2020] [Accepted: 02/07/2020] [Indexed: 12/11/2022] Open
Abstract
Rhabdomyosarcoma (RMS) and rhabdoid tumors (RT) are rare soft-tissue malignancies with the highest incidence in infants, children, and adolescents. Advanced, recurrent, and/or metastatic RMS and RT exhibit poor response to treatment. One of the main mechanisms behind resistance to treatment is believed to be intratumoral heterogeneity. In this study, we investigated the myogenic determination factor 1 (MYOD1) and Noggin (NOG) markers in an embryonal RMS (ERMS) cell line and an RT cell line and the differential response of the MYOD1 and NOG expressing subpopulations to chemotherapy. Importantly, we found that these markers together identify a subpopulation of cells (MYOD1+ NOG+ cells) with primary resistance to Vincristine and Doxorubicin, two commonly used chemotherapies for ERMS and RT. The chemoresistant MYOD1+ NOG+ cells express markers of undifferentiated cells such as myogenin and ID1. Combination of Vincristine with TPA/GSK126, a drug combination shown to induce differentiation of RMS cell lines, is able to partially overcome MYOD1/NOG cells chemoresistance.
Collapse
Affiliation(s)
| | | | | | - Richard D. Lackman
- Cooper University Hospital, Camden, NJ, USA
- MD Anderson Cancer Center at Cooper, Camden, NJ, USA
| | | | | | - Zeus A. Antonello
- Cooper University Hospital, Camden, NJ, USA
- Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Igor I. Kuzin
- Cooper University Hospital, Camden, NJ, USA
- Cooper Medical School of Rowan University, Camden, NJ, USA
| |
Collapse
|
30
|
La transplantation de cellules tumorales chez le poisson zèbre : de la recherche translationnelle à la médecine personnalisée. Bull Cancer 2020; 107:30-40. [DOI: 10.1016/j.bulcan.2019.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/24/2022]
|
31
|
Adhikari A, Davie J. Wnt deregulation in rhabdomyosarcoma. Stem Cell Investig 2019; 6:13. [PMID: 31304179 DOI: 10.21037/sci.2019.06.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/06/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Abhinav Adhikari
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine and Simmons Cancer Institute, Carbondale, IL, USA
| | - Judith Davie
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine and Simmons Cancer Institute, Carbondale, IL, USA
| |
Collapse
|
32
|
Yan C, Brunson DC, Tang Q, Do D, Iftimia NA, Moore JC, Hayes MN, Welker AM, Garcia EG, Dubash TD, Hong X, Drapkin BJ, Myers DT, Phat S, Volorio A, Marvin DL, Ligorio M, Dershowitz L, McCarthy KM, Karabacak MN, Fletcher JA, Sgroi DC, Iafrate JA, Maheswaran S, Dyson NJ, Haber DA, Rawls JF, Langenau DM. Visualizing Engrafted Human Cancer and Therapy Responses in Immunodeficient Zebrafish. Cell 2019; 177:1903-1914.e14. [PMID: 31031007 PMCID: PMC6570580 DOI: 10.1016/j.cell.2019.04.004] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 02/19/2019] [Accepted: 03/31/2019] [Indexed: 01/06/2023]
Abstract
Xenograft cell transplantation into immunodeficient mice has become the gold standard for assessing pre-clinical efficacy of cancer drugs, yet direct visualization of single-cell phenotypes is difficult. Here, we report an optically-clear prkdc-/-, il2rga-/- zebrafish that lacks adaptive and natural killer immune cells, can engraft a wide array of human cancers at 37°C, and permits the dynamic visualization of single engrafted cells. For example, photoconversion cell-lineage tracing identified migratory and proliferative cell states in human rhabdomyosarcoma, a pediatric cancer of muscle. Additional experiments identified the preclinical efficacy of combination olaparib PARP inhibitor and temozolomide DNA-damaging agent as an effective therapy for rhabdomyosarcoma and visualized therapeutic responses using a four-color FUCCI cell-cycle fluorescent reporter. These experiments identified that combination treatment arrested rhabdomyosarcoma cells in the G2 cell cycle prior to induction of apoptosis. Finally, patient-derived xenografts could be engrafted into our model, opening new avenues for developing personalized therapeutic approaches in the future.
Collapse
Affiliation(s)
- Chuan Yan
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Dalton C Brunson
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Bethesda, MD 20815, USA
| | - Qin Tang
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Bethesda, MD 20815, USA
| | - Daniel Do
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Bethesda, MD 20815, USA
| | - Nicolae A Iftimia
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - John C Moore
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Madeline N Hayes
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Alessandra M Welker
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Elaine G Garcia
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Taronish D Dubash
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Xin Hong
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Benjamin J Drapkin
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - David T Myers
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Sarah Phat
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Angela Volorio
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Dieuwke L Marvin
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Matteo Ligorio
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Lyle Dershowitz
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Karin M McCarthy
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Murat N Karabacak
- Shriners Hospitals for Children-Boston, MA 02114, USA; Center for Engineering in Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02114, USA
| | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Dennis C Sgroi
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - John A Iafrate
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Nick J Dyson
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Howard Hughes Medical Institute, Bethesda, MD 20815, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - David M Langenau
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
33
|
Hayes MN, McCarthy K, Jin A, Oliveira ML, Iyer S, Garcia SP, Sindiri S, Gryder B, Motala Z, Nielsen GP, Borg JP, van de Rijn M, Malkin D, Khan J, Ignatius MS, Langenau DM. Vangl2/RhoA Signaling Pathway Regulates Stem Cell Self-Renewal Programs and Growth in Rhabdomyosarcoma. Cell Stem Cell 2019; 22:414-427.e6. [PMID: 29499154 DOI: 10.1016/j.stem.2018.02.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 12/14/2017] [Accepted: 02/06/2018] [Indexed: 01/09/2023]
Abstract
Tumor growth and relapse are driven by tumor propagating cells (TPCs). However, mechanisms regulating TPC fate choices, maintenance, and self-renewal are not fully understood. Here, we show that Van Gogh-like 2 (Vangl2), a core regulator of the non-canonical Wnt/planar cell polarity (Wnt/PCP) pathway, affects TPC self-renewal in rhabdomyosarcoma (RMS)-a pediatric cancer of muscle. VANGL2 is expressed in a majority of human RMS and within early mononuclear progenitor cells. VANGL2 depletion inhibited cell proliferation, reduced TPC numbers, and induced differentiation of human RMS in vitro and in mouse xenografts. Using a zebrafish model of embryonal rhabdomyosarcoma (ERMS), we determined that Vangl2 expression enriches for TPCs and promotes their self-renewal. Expression of constitutively active and dominant-negative isoforms of RHOA revealed that it acts downstream of VANGL2 to regulate proliferation and maintenance of TPCs in human RMS. Our studies offer insights into pathways that control TPCs and identify new potential therapeutic targets.
Collapse
Affiliation(s)
- Madeline N Hayes
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital Research Institute, Boston, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Karin McCarthy
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital Research Institute, Boston, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Alexander Jin
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital Research Institute, Boston, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Mariana L Oliveira
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital Research Institute, Boston, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Sowmya Iyer
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital Research Institute, Boston, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Sara P Garcia
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital Research Institute, Boston, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Sivasish Sindiri
- Oncogenomics Section, Center for Cancer Research, National Cancer Institute, NIH, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Berkley Gryder
- Oncogenomics Section, Center for Cancer Research, National Cancer Institute, NIH, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Zainab Motala
- Division of Hematology/Oncology, Hospital for Sick Children and Department of Pediatrics, University of Toronto, Toronto, ON M5G1X8, Canada
| | - G Petur Nielsen
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Jean-Paul Borg
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Inst Paoli Calmettes, UMR7258 CNRS, U1068 INSERM, "Cell Polarity, Cell signalling and Cancer - Equipe labellisée Ligue Contre le Cancer," Marseille, France
| | - Matt van de Rijn
- Department of Pathology, Stanford University Medical Center, Stanford, CA 94305, USA
| | - David Malkin
- Division of Hematology/Oncology, Hospital for Sick Children and Department of Pediatrics, University of Toronto, Toronto, ON M5G1X8, Canada
| | - Javed Khan
- Oncogenomics Section, Center for Cancer Research, National Cancer Institute, NIH, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Myron S Ignatius
- Molecular Medicine and Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX 78229, USA
| | - David M Langenau
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital Research Institute, Boston, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
34
|
Talbot JC, Teets EM, Ratnayake D, Duy PQ, Currie PD, Amacher SL. Muscle precursor cell movements in zebrafish are dynamic and require Six family genes. Development 2019; 146:dev171421. [PMID: 31023879 PMCID: PMC6550023 DOI: 10.1242/dev.171421] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/16/2019] [Indexed: 01/09/2023]
Abstract
Muscle precursors need to be correctly positioned during embryonic development for proper body movement. In zebrafish, a subset of hypaxial muscle precursors from the anterior somites undergo long-range migration, moving away from the trunk in three streams to form muscles in distal locations such as the fin. We mapped long-distance muscle precursor migrations with unprecedented resolution using live imaging. We identified conserved genes necessary for normal precursor motility (six1a, six1b, six4a, six4b and met). These genes are required for movement away from somites and later to partition two muscles within the fin bud. During normal development, the middle muscle precursor stream initially populates the fin bud, then the remainder of this stream contributes to the posterior hypaxial muscle. When we block fin bud development by impairing retinoic acid synthesis or Fgfr function, the entire stream contributes to the posterior hypaxial muscle indicating that muscle precursors are not committed to the fin during migration. Our findings demonstrate a conserved muscle precursor motility pathway, identify dynamic cell movements that generate posterior hypaxial and fin muscles, and demonstrate flexibility in muscle precursor fates.
Collapse
Affiliation(s)
- Jared C Talbot
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
| | - Emily M Teets
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Dhanushika Ratnayake
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- EMBL Australia, Monash University, Clayton, VIC, 3800, Australia
| | - Phan Q Duy
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- EMBL Australia, Monash University, Clayton, VIC, 3800, Australia
| | - Sharon L Amacher
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
- Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
35
|
How Surrogate and Chemical Genetics in Model Organisms Can Suggest Therapies for Human Genetic Diseases. Genetics 2018; 208:833-851. [PMID: 29487144 PMCID: PMC5844338 DOI: 10.1534/genetics.117.300124] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/26/2017] [Indexed: 12/12/2022] Open
Abstract
Genetic diseases are both inherited and acquired. Many genetic diseases fall under the paradigm of orphan diseases, a disease found in < 1 in 2000 persons. With rapid and cost-effective genome sequencing becoming the norm, many causal mutations for genetic diseases are being rapidly determined. In this regard, model organisms are playing an important role in validating if specific mutations identified in patients drive the observed phenotype. An emerging challenge for model organism researchers is the application of genetic and chemical genetic platforms to discover drug targets and drugs/drug-like molecules for potential treatment options for patients with genetic disease. This review provides an overview of how model organisms have contributed to our understanding of genetic disease, with a focus on the roles of yeast and zebrafish in gene discovery and the identification of compounds that could potentially treat human genetic diseases.
Collapse
|
36
|
Ignatius MS, Hayes MN, Moore FE, Tang Q, Garcia SP, Blackburn PR, Baxi K, Wang L, Jin A, Ramakrishnan A, Reeder S, Chen Y, Nielsen GP, Chen EY, Hasserjian RP, Tirode F, Ekker SC, Langenau DM. tp53 deficiency causes a wide tumor spectrum and increases embryonal rhabdomyosarcoma metastasis in zebrafish. eLife 2018; 7:37202. [PMID: 30192230 PMCID: PMC6128690 DOI: 10.7554/elife.37202] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/22/2018] [Indexed: 12/22/2022] Open
Abstract
The TP53 tumor-suppressor gene is mutated in >50% of human tumors and Li-Fraumeni patients with germ line inactivation are predisposed to developing cancer. Here, we generated tp53 deleted zebrafish that spontaneously develop malignant peripheral nerve-sheath tumors, angiosarcomas, germ cell tumors, and an aggressive Natural Killer cell-like leukemia for which no animal model has been developed. Because the tp53 deletion was generated in syngeneic zebrafish, engraftment of fluorescent-labeled tumors could be dynamically visualized over time. Importantly, engrafted tumors shared gene expression signatures with predicted cells of origin in human tissue. Finally, we showed that tp53del/del enhanced invasion and metastasis in kRASG12D-induced embryonal rhabdomyosarcoma (ERMS), but did not alter the overall frequency of cancer stem cells, suggesting novel pro-metastatic roles for TP53 loss-of-function in human muscle tumors. In summary, we have developed a Li-Fraumeni zebrafish model that is amenable to large-scale transplantation and direct visualization of tumor growth in live animals.
Collapse
Affiliation(s)
- Myron S Ignatius
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts.,Center of Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts.,Harvard Stem Cell Institute, Boston, Massachusetts.,Department of Molecular Medicine, Greehey Children's Cancer Research Institute, San Antonio, Texas
| | - Madeline N Hayes
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts.,Center of Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts.,Harvard Stem Cell Institute, Boston, Massachusetts
| | - Finola E Moore
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts.,Center of Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts.,Harvard Stem Cell Institute, Boston, Massachusetts
| | - Qin Tang
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts.,Center of Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts.,Harvard Stem Cell Institute, Boston, Massachusetts
| | - Sara P Garcia
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts
| | - Patrick R Blackburn
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, United States
| | - Kunal Baxi
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute, San Antonio, Texas
| | - Long Wang
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute, San Antonio, Texas
| | - Alexander Jin
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts
| | - Ashwin Ramakrishnan
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts
| | - Sophia Reeder
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts
| | - Yidong Chen
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute, San Antonio, Texas
| | - Gunnlaugur Petur Nielsen
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts.,Center of Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts
| | - Eleanor Y Chen
- Department of Pathology, University of Washington, Seattle, United States
| | - Robert P Hasserjian
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts.,Center of Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts
| | - Franck Tirode
- Department of Translational Research and Innovation, Université Claude Bernard Lyon, Cancer Research Center of Lyon, Lyon, France
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - David M Langenau
- Department of Pathology, Massachusetts General Hospital Research Institute, Boston, Massachusetts.,Center of Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts.,Harvard Stem Cell Institute, Boston, Massachusetts
| |
Collapse
|
37
|
Codenotti S, Poli M, Asperti M, Zizioli D, Marampon F, Fanzani A. Cell growth potential drives ferroptosis susceptibility in rhabdomyosarcoma and myoblast cell lines. J Cancer Res Clin Oncol 2018; 144:1717-1730. [PMID: 29971532 DOI: 10.1007/s00432-018-2699-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 06/29/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE Ferroptosis is a programmed form of iron-dependent cell death caused by lipid hydroperoxide accumulation, which can be prevented by glutathione peroxidase 4 (GPx4) activity. Here we investigated the effects of ferroptosis inducers called erastin and RSL3, which act by glutathione depletion and GPx4 inactivation, respectively, on muscle-derived cell lines of embryonal and alveolar rhabdomyosarcoma (RMS), and mouse normal skeletal C2C12 myoblasts. METHODS Myogenic lines were exposed to stepwise increasing concentrations of ferroptosis inducers either alone or in combination with iron supplementation, iron chelating agents (bathophenanthrolinedisulfonic acid, BPS), antioxidant molecules (glutathione, N-acetylcysteine), lipid peroxidation inhibitors (ferrostatin-1), and chemotherapeutic agents (doxorubicin and actinomycin D). Drug susceptibility was quantified by measuring cell viability, proliferation and differentiation via neutral red assay, crystal violet assay and Giemsa staining, respectively. The detection of lipid hydroperoxide and protein levels was performed by immunofluorescence and Western blot analysis, respectively. RESULTS Erastin and RSL3 increased lipid hydroperoxide levels preferentially in the embryonal U57810 and myoblast C2C12 lines, leading to ferroptosis that was accentuated by iron supplementation or prevented by co-treatment with BPS, glutathione, N-acetylcysteine and ferrostatin-1. The inhibition of extracellular regulated kinases (ERK) pathway prevented ferroptosis in U57810 and C2C12 cells, whereas its increased activation in the embryonal RD cells mediated by caveolin-1 (Cav-1) overexpression led to augmented ferroptosis susceptibility. Finally, we observed the combination of erastin or RSL3 with chemotherapeutic doxorubicin and actinomycin D agents to be effective in increasing cell death in all RMS lines. CONCLUSIONS Erastin and RSL3 trigger ferroptosis in highly proliferating myogenic lines through a ERK pathway-dependent fashion.
Collapse
Affiliation(s)
- Silvia Codenotti
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maura Poli
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Michela Asperti
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Zizioli
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesco Marampon
- Division of Radiation Oncology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Fanzani
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
38
|
Kartha VK, Alamoud KA, Sadykov K, Nguyen BC, Laroche F, Feng H, Lee J, Pai SI, Varelas X, Egloff AM, Snyder-Cappione JE, Belkina AC, Bais MV, Monti S, Kukuruzinska MA. Functional and genomic analyses reveal therapeutic potential of targeting β-catenin/CBP activity in head and neck cancer. Genome Med 2018; 10:54. [PMID: 30029671 PMCID: PMC6053793 DOI: 10.1186/s13073-018-0569-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/11/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy characterized by tumor heterogeneity, locoregional metastases, and resistance to existing treatments. Although a number of genomic and molecular alterations associated with HNSCC have been identified, they have had limited impact on the clinical management of this disease. To date, few targeted therapies are available for HNSCC, and only a small fraction of patients have benefited from these treatments. A frequent feature of HNSCC is the inappropriate activation of β-catenin that has been implicated in cell survival and in the maintenance and expansion of stem cell-like populations, thought to be the underlying cause of tumor recurrence and resistance to treatment. However, the therapeutic value of targeting β-catenin activity in HNSCC has not been explored. METHODS We utilized a combination of computational and experimental profiling approaches to examine the effects of blocking the interaction between β-catenin and cAMP-responsive element binding (CREB)-binding protein (CBP) using the small molecule inhibitor ICG-001. We generated and annotated in vitro treatment gene expression signatures of HNSCC cells, derived from human oral squamous cell carcinomas (OSCCs), using microarrays. We validated the anti-tumorigenic activity of ICG-001 in vivo using SCC-derived tumor xenografts in murine models, as well as embryonic zebrafish-based screens of sorted stem cell-like subpopulations. Additionally, ICG-001-inhibition signatures were overlaid with RNA-sequencing data from The Cancer Genome Atlas (TCGA) for human OSCCs to evaluate its association with tumor progression and prognosis. RESULTS ICG-001 inhibited HNSCC cell proliferation and tumor growth in cellular and murine models, respectively, while promoting intercellular adhesion and loss of invasive phenotypes. Furthermore, ICG-001 preferentially targeted the ability of subpopulations of stem-like cells to establish metastatic tumors in zebrafish. Significantly, interrogation of the ICG-001 inhibition-associated gene expression signature in the TCGA OSCC human cohort indicated that the targeted β-catenin/CBP transcriptional activity tracked with tumor status, advanced tumor grade, and poor overall patient survival. CONCLUSIONS Collectively, our results identify β-catenin/CBP interaction as a novel target for anti-HNSCC therapy and provide evidence that derivatives of ICG-001 with enhanced inhibitory activity may serve as an effective strategy to interfere with aggressive features of HNSCC.
Collapse
Affiliation(s)
- Vinay K Kartha
- Bioinformatics Program, Boston University, Boston, MA, USA
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Khalid A Alamoud
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA
| | - Khikmet Sadykov
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA
| | - Bach-Cuc Nguyen
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA
| | - Fabrice Laroche
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Hui Feng
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Jina Lee
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara I Pai
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Ann Marie Egloff
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jennifer E Snyder-Cappione
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Anna C Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| | - Manish V Bais
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA
| | - Stefano Monti
- Bioinformatics Program, Boston University, Boston, MA, USA
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Maria A Kukuruzinska
- Department of Molecular and Cell Biology, Goldman School of Dental Medicine, Boston University School of Medicine, 72 East Concord Street, E4, Boston, MA, 02118, USA.
| |
Collapse
|
39
|
SNAIL is a key regulator of alveolar rhabdomyosarcoma tumor growth and differentiation through repression of MYF5 and MYOD function. Cell Death Dis 2018; 9:643. [PMID: 29844345 PMCID: PMC5974324 DOI: 10.1038/s41419-018-0693-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/30/2018] [Accepted: 05/10/2018] [Indexed: 12/13/2022]
Abstract
Rhabdomyosarcoma (RMS) is a mesenchymal tumor of soft tissue in children that originates from a myogenic differentiation defect. Expression of SNAIL transcription factor is elevated in the alveolar subtype of RMS (ARMS), characterized by a low myogenic differentiation status and high aggressiveness. In RMS patients SNAIL level increases with higher stage. Moreover, SNAIL level negatively correlates with MYF5 expression. The differentiation of human ARMS cells diminishes SNAIL level. SNAIL silencing in ARMS cells inhibits proliferation and induces differentiation in vitro, and thereby completely abolishes the growth of human ARMS xenotransplants in vivo. SNAIL silencing induces myogenic differentiation by upregulation of myogenic factors and muscle-specific microRNAs, such as miR-206. SNAIL binds to the MYF5 promoter suppressing its expression. SNAIL displaces MYOD from E-box sequences (CANNTG) that are associated with genes expressed during differentiation and G/C rich in their central dinucleotides. SNAIL silencing allows the re-expression of MYF5 and canonical MYOD binding, promoting ARMS cell myogenic differentiation. In differentiating ARMS cells SNAIL forms repressive complex with histone deacetylates 1 and 2 (HDAC1/2) and regulates their expression. Accordingly, in human myoblasts SNAIL silencing induces differentiation by upregulation of myogenic factors. Our data clearly point to SNAIL as a key regulator of myogenic differentiation and a new promising target for future ARMS therapies.
Collapse
|
40
|
Völkel P, Dupret B, Le Bourhis X, Angrand PO. [The zebrafish model in oncology]. Med Sci (Paris) 2018; 34:345-353. [PMID: 29658479 DOI: 10.1051/medsci/20183404016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Although cell culture and mouse models will remain a cornerstone of cancer research, the unique capabilities of the zebrafish outline the potential of this model for shedding light on cancer biology in vivo. Zebrafish develops cancers spontaneously, after chemical mutagenesis or through genetic manipulations. Furthermore, zebrafish cancers are similar to human tumors at the histological and molecular levels allowing the study of tumor initiation, progression and heterogeneity. Xenotransplantation of human cancer cells in embryos or adult zebrafish presents the advantage of following cancer cell behavior in vivo. Finally, zebrafish embryos are used in molecule screens and contribute to the identification of novel anti-cancer therapeutic strategies. Here, we review different involvements of the zebrafish model in cancer research.
Collapse
Affiliation(s)
- Pamela Völkel
- CNRS Lille, Inserm U908, Université de Lille, Bâtiment SN3, Cité Scientifique, 59655 Villeneuve d'Ascq, France
| | - Babara Dupret
- Inserm U908, Université de Lille, Bâtiment SN3, Cité Scientifique, 59655 Villeneuve d'Ascq, France
| | - Xuefen Le Bourhis
- Inserm U908, Université de Lille, Bâtiment SN3, Cité Scientifique, 59655 Villeneuve d'Ascq, France
| | - Pierre-Olivier Angrand
- Inserm U908, Université de Lille, Bâtiment SN3, Cité Scientifique, 59655 Villeneuve d'Ascq, France
| |
Collapse
|
41
|
Ignatius MS, Hayes MN, Lobbardi R, Chen EY, McCarthy KM, Sreenivas P, Motala Z, Durbin AD, Molodtsov A, Reeder S, Jin A, Sindiri S, Beleyea BC, Bhere D, Alexander MS, Shah K, Keller C, Linardic CM, Nielsen PG, Malkin D, Khan J, Langenau DM. The NOTCH1/SNAIL1/MEF2C Pathway Regulates Growth and Self-Renewal in Embryonal Rhabdomyosarcoma. Cell Rep 2018; 19:2304-2318. [PMID: 28614716 PMCID: PMC5563075 DOI: 10.1016/j.celrep.2017.05.061] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/08/2017] [Accepted: 05/17/2017] [Indexed: 12/27/2022] Open
Abstract
Tumor-propagating cells (TPCs) share self-renewal properties with normal stem cells and drive continued tumor growth. However, mechanisms regulating TPC self-renewal are largely unknown, especially in embryonal rhabdomyosarcoma (ERMS)-a common pediatric cancer of muscle. Here, we used a zebrafish transgenic model of ERMS to identify a role for intracellular NOTCH1 (ICN1) in increasing TPCs by 23-fold. ICN1 expanded TPCs by enabling the de-differentiation of zebrafish ERMS cells into self-renewing myf5+ TPCs, breaking the rigid differentiation hierarchies reported in normal muscle. ICN1 also had conserved roles in regulating human ERMS self-renewal and growth. Mechanistically, ICN1 upregulated expression of SNAIL1, a transcriptional repressor, to increase TPC number in human ERMS and to block muscle differentiation through suppressing MEF2C, a myogenic differentiation transcription factor. Our data implicate the NOTCH1/SNAI1/MEF2C signaling axis as a major determinant of TPC self-renewal and differentiation in ERMS, raising hope of therapeutically targeting this pathway in the future.
Collapse
Affiliation(s)
- Myron S Ignatius
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA; Greehey Children's Cancer Research Institute and Department of Molecular Medicine, UT Health Sciences Center, San Antonio, TX 78229, USA
| | - Madeline N Hayes
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA
| | - Riadh Lobbardi
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA
| | - Eleanor Y Chen
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Karin M McCarthy
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Prethish Sreenivas
- Greehey Children's Cancer Research Institute and Department of Molecular Medicine, UT Health Sciences Center, San Antonio, TX 78229, USA
| | - Zainab Motala
- Division of Hematology/Oncology, Hospital for Sick Children and Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Adam D Durbin
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02215, USA
| | - Aleksey Molodtsov
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sophia Reeder
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alexander Jin
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sivasish Sindiri
- Oncogenomics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Brian C Beleyea
- Department of Pediatrics and Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Deepak Bhere
- Molecular Neurotherapy and Imaging Laboratory, Stem Cell Therapeutics and Imaging Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Matthew S Alexander
- Department of Pediatrics and Genetics, Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Khalid Shah
- Department of Pediatrics and Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Charles Keller
- Children's Cancer Therapy Development Institute, Beaverton, OR 97005, USA
| | - Corinne M Linardic
- Oncogenomics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Petur G Nielsen
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - David Malkin
- Division of Hematology/Oncology, Hospital for Sick Children and Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Javed Khan
- Oncogenomics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - David M Langenau
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA; Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Stem Cell Institute, Boston, MA 02114, USA.
| |
Collapse
|
42
|
Mizgirev IV, Safina DR, Demidyuk IV, Kostrov SV. Organism-Level Tumor Models in Zebrafish Danio rerio. Acta Naturae 2018; 10:24-29. [PMID: 30116612 PMCID: PMC6087818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Indexed: 11/18/2022] Open
Abstract
Development and implementation of adequate organism-level models is one of the key elements in biomedical research that focuses on experimental oncology. Over the last decade, studies using Zebrafish (Danio rerio) have gained in popularity in this area of research. This review describes the various approaches that have been used in developing highly effective models for oncological (clinical term, better cancer or tumor) studies based on D. rerio. Priority is given to transplantation models of cancer and their application to optically transparent D. rerio lines, including clonal ones, and utilization tumors of various origins bearing fluorescent labels. The combination of tumor transplantation at organism-level models in transparent clonal D. rerio lines with fluorescent microscopy, FACS-fractionation of tumor cell subsets, and transcription analysis can result in one of the most promising research approaches in providing new information on tumor formation and growth.
Collapse
Affiliation(s)
- I. V. Mizgirev
- N.N. Petrov National Medical Research Center of Oncology, Ministry of Health of Russia, Leningradskaya Str., 68, Pesochnyy Settlement, St. Petersburg, 197758, Russia
| | - D. R. Safina
- Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq., 2, Moscow, 123182 , Russia
| | - I. V. Demidyuk
- Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq., 2, Moscow, 123182 , Russia
| | - S. V. Kostrov
- Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq., 2, Moscow, 123182 , Russia
| |
Collapse
|
43
|
Deel MD, Slemmons KK, Hinson AR, Genadry KC, Burgess BA, Crose LES, Kuprasertkul N, Oristian KM, Bentley RC, Linardic CM. The Transcriptional Coactivator TAZ Is a Potent Mediator of Alveolar Rhabdomyosarcoma Tumorigenesis. Clin Cancer Res 2018. [PMID: 29514840 DOI: 10.1158/1078-0432.ccr-17-1207] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: Alveolar rhabdomyosarcoma (aRMS) is a childhood soft tissue sarcoma driven by the signature PAX3-FOXO1 (P3F) fusion gene. Five-year survival for aRMS is <50%, with no improvement in over 4 decades. Although the transcriptional coactivator TAZ is oncogenic in carcinomas, the role of TAZ in sarcomas is poorly understood. The aim of this study was to investigate the role of TAZ in P3F-aRMS tumorigenesis.Experimental Design: After determining from publicly available datasets that TAZ is upregulated in human aRMS transcriptomes, we evaluated whether TAZ is also upregulated in our myoblast-based model of P3F-initiated tumorigenesis, and performed IHC staining of 63 human aRMS samples from tissue microarrays. Using constitutive and inducible RNAi, we examined the impact of TAZ loss of function on aRMS oncogenic phenotypes in vitro and tumorigenesis in vivo Finally, we performed pharmacologic studies in aRMS cell lines using porphyrin compounds, which interfere with TAZ-TEAD transcriptional activity.Results: TAZ is upregulated in our P3F-initiated aRMS model, and aRMS cells and tumors have high nuclear TAZ expression. In vitro, TAZ suppression inhibits aRMS cell proliferation, induces apoptosis, supports myogenic differentiation, and reduces aRMS cell stemness. TAZ-deficient aRMS cells are enriched in G2-M phase of the cell cycle. In vivo, TAZ suppression attenuates aRMS xenograft tumor growth. Preclinical studies show decreased aRMS xenograft tumor growth with porphyrin compounds alone and in combination with vincristine.Conclusions: TAZ is oncogenic in aRMS sarcomagenesis. While P3F is currently not therapeutically tractable, targeting TAZ could be a promising novel approach in aRMS. Clin Cancer Res; 24(11); 2616-30. ©2018 AACR.
Collapse
Affiliation(s)
- Michael D Deel
- Division of Hematology-Oncology, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina
| | - Katherine K Slemmons
- Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Ashley R Hinson
- Division of Hematology-Oncology, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina
| | - Katia C Genadry
- Division of Hematology-Oncology, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina
| | - Breanne A Burgess
- Division of Hematology-Oncology, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina
| | - Lisa E S Crose
- Division of Hematology-Oncology, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina
| | | | - Kristianne M Oristian
- Division of Hematology-Oncology, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina
| | - Rex C Bentley
- Department of Pathology, School of Medicine, Duke University, Durham, North Carolina
| | - Corinne M Linardic
- Division of Hematology-Oncology, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina. .,Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, North Carolina
| |
Collapse
|
44
|
Habbsa S, McKinstry M, Bowman TV. “Sea”-ing Is Believing: In Vivo Imaging of Hematopoietic Stem Cells and Cancer Using Zebrafish. CURRENT STEM CELL REPORTS 2017. [DOI: 10.1007/s40778-017-0088-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Mohamed A, Sun C, De Mello V, Selfe J, Missiaglia E, Shipley J, Murray GI, Zammit PS, Wackerhage H. The Hippo effector TAZ (WWTR1) transforms myoblasts and TAZ abundance is associated with reduced survival in embryonal rhabdomyosarcoma. J Pathol 2017; 240:3-14. [PMID: 27184927 PMCID: PMC4995731 DOI: 10.1002/path.4745] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 04/04/2016] [Accepted: 04/27/2016] [Indexed: 12/22/2022]
Abstract
The Hippo effector YAP has recently been identified as a potent driver of embryonal rhabdomyosarcoma (ERMS). Most reports suggest that the YAP paralogue TAZ (gene symbol WWTR1) functions as YAP but, in skeletal muscle, TAZ has been reported to promote myogenic differentiation, whereas YAP inhibits it. Here, we investigated whether TAZ is also a rhabdomyosarcoma oncogene or whether TAZ acts as a YAP antagonist. Immunostaining of rhabdomyosarcoma tissue microarrays revealed that TAZ is significantly associated with poor survival in ERMS. In 12% of fusion gene‐negative rhabdomyosarcomas, the TAZ locus is gained, which is correlated with increased expression. Constitutively active TAZ S89A significantly increased proliferation of C2C12 myoblasts and, importantly, colony formation on soft agar, suggesting transformation. However, TAZ then switches to enhance myogenic differentiation in C2C12 myoblasts, unlike YAP. Conversely, lentiviral shRNA‐mediated TAZ knockdown in human ERMS cells reduced proliferation and anchorage‐independent growth. While TAZ S89A or YAP1 S127A similarly activated the 8XGTIIC–Luc Hippo reporter, only YAP1 S127A activated the Brachyury (T‐box) reporter. Consistent with its oncogene function, TAZ S89A induced expression of the ERMS cancer stem cell gene Myf5 and the serine biosynthesis pathway (Phgdh, Psat1, Psph) in C2C12 myoblasts. Thus, TAZ is associated with poor survival in ERMS and could act as an oncogene in rhabdomyosarcoma. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Abdalla Mohamed
- School of Medicine, Dentistry and Nutrition, University of Aberdeen, UK
| | - Congshan Sun
- Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Vanessa De Mello
- School of Medicine, Dentistry and Nutrition, University of Aberdeen, UK
| | - Joanna Selfe
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, UK
| | | | - Janet Shipley
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, UK
| | - Graeme I Murray
- School of Medicine, Dentistry and Nutrition, University of Aberdeen, UK
| | - Pete S Zammit
- Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | | |
Collapse
|
46
|
Berberoglu MA, Gallagher TL, Morrow ZT, Talbot JC, Hromowyk KJ, Tenente IM, Langenau DM, Amacher SL. Satellite-like cells contribute to pax7-dependent skeletal muscle repair in adult zebrafish. Dev Biol 2017; 424:162-180. [PMID: 28279710 DOI: 10.1016/j.ydbio.2017.03.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/02/2017] [Accepted: 03/05/2017] [Indexed: 12/24/2022]
Abstract
Satellite cells, also known as muscle stem cells, are responsible for skeletal muscle growth and repair in mammals. Pax7 and Pax3 transcription factors are established satellite cell markers required for muscle development and regeneration, and there is great interest in identifying additional factors that regulate satellite cell proliferation, differentiation, and/or skeletal muscle regeneration. Due to the powerful regenerative capacity of many zebrafish tissues, even in adults, we are exploring the regenerative potential of adult zebrafish skeletal muscle. Here, we show that adult zebrafish skeletal muscle contains cells similar to mammalian satellite cells. Adult zebrafish satellite-like cells have dense heterochromatin, express Pax7 and Pax3, proliferate in response to injury, and show peak myogenic responses 4-5 days post-injury (dpi). Furthermore, using a pax7a-driven GFP reporter, we present evidence implicating satellite-like cells as a possible source of new muscle. In lieu of central nucleation, which distinguishes regenerating myofibers in mammals, we describe several characteristics that robustly identify newly-forming myofibers from surrounding fibers in injured adult zebrafish muscle. These characteristics include partially overlapping expression in satellite-like cells and regenerating myofibers of two RNA-binding proteins Rbfox2 and Rbfoxl1, known to regulate embryonic muscle development and function. Finally, by analyzing pax7a; pax7b double mutant zebrafish, we show that Pax7 is required for adult skeletal muscle repair, as it is in the mouse.
Collapse
Affiliation(s)
- Michael A Berberoglu
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
| | - Thomas L Gallagher
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
| | - Zachary T Morrow
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
| | - Jared C Talbot
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
| | - Kimberly J Hromowyk
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
| | - Inês M Tenente
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Department of Molecular Pathology and Regenerative Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - David M Langenau
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Department of Molecular Pathology and Regenerative Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Sharon L Amacher
- Departments of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA.
| |
Collapse
|
47
|
Disruption of myoblast alignment by highly motile rhabdomyosarcoma cell in tissue structure. J Biosci Bioeng 2017; 123:259-264. [DOI: 10.1016/j.jbiosc.2016.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/01/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022]
|
48
|
Tenente IM, Hayes MN, Ignatius MS, McCarthy K, Yohe M, Sindiri S, Gryder B, Oliveira ML, Ramakrishnan A, Tang Q, Chen EY, Petur Nielsen G, Khan J, Langenau DM. Myogenic regulatory transcription factors regulate growth in rhabdomyosarcoma. eLife 2017; 6. [PMID: 28080960 PMCID: PMC5231408 DOI: 10.7554/elife.19214] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 12/08/2016] [Indexed: 01/01/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a pediatric malignacy of muscle with myogenic regulatory transcription factors MYOD and MYF5 being expressed in this disease. Consensus in the field has been that expression of these factors likely reflects the target cell of transformation rather than being required for continued tumor growth. Here, we used a transgenic zebrafish model to show that Myf5 is sufficient to confer tumor-propagating potential to RMS cells and caused tumors to initiate earlier and have higher penetrance. Analysis of human RMS revealed that MYF5 and MYOD are mutually-exclusively expressed and each is required for sustained tumor growth. ChIP-seq and mechanistic studies in human RMS uncovered that MYF5 and MYOD bind common DNA regulatory elements to alter transcription of genes that regulate muscle development and cell cycle progression. Our data support unappreciated and dominant oncogenic roles for MYF5 and MYOD convergence on common transcriptional targets to regulate human RMS growth. DOI:http://dx.doi.org/10.7554/eLife.19214.001
Collapse
Affiliation(s)
- Inês M Tenente
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States.,GABBA Program, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - Madeline N Hayes
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| | - Myron S Ignatius
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States.,Molecular Medicine, Greehey Children's Cancer Research Institute, San Antonio, United States
| | - Karin McCarthy
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| | - Marielle Yohe
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, Gaithersburg, United States
| | - Sivasish Sindiri
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, Gaithersburg, United States
| | - Berkley Gryder
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, Gaithersburg, United States
| | - Mariana L Oliveira
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ashwin Ramakrishnan
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| | - Qin Tang
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| | - Eleanor Y Chen
- Department of Pathology, University of Washington, Seattle, United States
| | - G Petur Nielsen
- Department of Pathology, Massachusetts General Hospital, Boston, United States
| | - Javed Khan
- Oncogenomics Section, Pediatric Oncology Branch, Advanced Technology Center, National Cancer Institute, Gaithersburg, United States
| | - David M Langenau
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, United States.,Harvard Stem Cell Institute, Cambridge, United States
| |
Collapse
|
49
|
Hayes MN, Langenau DM. Discovering novel oncogenic pathways and new therapies using zebrafish models of sarcoma. Methods Cell Biol 2017; 138:525-561. [PMID: 28129857 DOI: 10.1016/bs.mcb.2016.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Sarcoma is a type of cancer affecting connective, supportive, or soft tissue of mesenchymal origin. Despite rare incidence in adults (<1%), over 15% of pediatric cancers are sarcoma. Sadly, both adults and children with relapsed or metastatic disease have devastatingly high rates of mortality. Current treatment options for sarcoma include surgery, radiation, and/or chemotherapy; however, significant limitations exist with respect to the efficacy of these strategies. Strong impetus has been placed on the development of novel therapies and preclinical models for uncovering mechanisms involved in the development, progression, and therapy resistance of sarcoma. Over the past 15 years, the zebrafish has emerged as a powerful genetic model of human cancer. High genetic conservation when combined with a unique susceptibility to develop sarcoma has made the zebrafish an effective tool for studying these diseases. Transgenic and gene-activation strategies have been employed to develop zebrafish models of rhabdomyosarcoma, malignant peripheral nerve sheath tumors, Ewing's sarcoma, chordoma, hemangiosarcoma, and liposarcoma. These models all display remarkable molecular and histopathological conservation with their human cancer counterparts and have offered excellent platforms for understanding disease progression in vivo. Short tumor latency and the amenability of zebrafish for ex vivo manipulation, live imaging studies, and tumor cell transplantation have allowed for efficient study of sarcoma initiation, growth, self-renewal, and maintenance. When coupled with facile chemical genetic approaches, zebrafish models of sarcoma have provided a strong translational tool to uncover novel drug pathways and new therapeutic strategies.
Collapse
Affiliation(s)
- M N Hayes
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts General Hospital, Charlestown, MA, United States; Harvard Stem Cell Institute, Boston, MA, United States
| | - D M Langenau
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts General Hospital, Charlestown, MA, United States; Harvard Stem Cell Institute, Boston, MA, United States
| |
Collapse
|
50
|
Moore JC, Tang Q, Yordán NT, Moore FE, Garcia EG, Lobbardi R, Ramakrishnan A, Marvin DL, Anselmo A, Sadreyev RI, Langenau DM. Single-cell imaging of normal and malignant cell engraftment into optically clear prkdc-null SCID zebrafish. J Exp Med 2016; 213:2575-2589. [PMID: 27810924 PMCID: PMC5110017 DOI: 10.1084/jem.20160378] [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: 03/14/2016] [Accepted: 09/16/2016] [Indexed: 12/03/2022] Open
Abstract
Cell transplantation into immunodeficient mice has revolutionized our understanding of regeneration, stem cell self-renewal, and cancer; yet models for direct imaging of engrafted cells has been limited. Here, we characterize zebrafish with mutations in recombination activating gene 2 (rag2), DNA-dependent protein kinase (prkdc), and janus kinase 3 (jak3). Histology, RNA sequencing, and single-cell transcriptional profiling of blood showed that rag2 hypomorphic mutant zebrafish lack T cells, whereas prkdc deficiency results in loss of mature T and B cells and jak3 in T and putative Natural Killer cells. Although all mutant lines engraft fluorescently labeled normal and malignant cells, only the prkdc mutant fish reproduced as homozygotes and also survived injury after cell transplantation. Engraftment into optically clear casper, prkdc-mutant zebrafish facilitated dynamic live cell imaging of muscle regeneration, repopulation of muscle stem cells within their endogenous niche, and muscle fiber fusion at single-cell resolution. Serial imaging approaches also uncovered stochasticity in fluorescently labeled leukemia regrowth after competitive cell transplantation into prkdc mutant fish, providing refined models to assess clonal dominance and progression in the zebrafish. Our experiments provide an optimized and facile transplantation model, the casper, prkdc mutant zebrafish, for efficient engraftment and direct visualization of fluorescently labeled normal and malignant cells at single-cell resolution.
Collapse
Affiliation(s)
- John C Moore
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Qin Tang
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Nora Torres Yordán
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard University, Cambridge, MA 02138
| | - Finola E Moore
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Elaine G Garcia
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Riadh Lobbardi
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Ashwin Ramakrishnan
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Dieuwke L Marvin
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
| | - Anthony Anselmo
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - David M Langenau
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| |
Collapse
|