Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Pediatr Blood Cancer ; 66(10): e27869, 2019 10.
Article in English | MEDLINE | ID: mdl-31222885

ABSTRACT

Overall survival rates for pediatric patients with high-risk or relapsed rhabdomyosarcoma (RMS) have not improved significantly since the 1980s. Recent studies have identified a number of targetable vulnerabilities in RMS, but these discoveries have infrequently translated into clinical trials. We propose streamlining the process by which agents are selected for clinical evaluation in RMS. We believe that strong consideration should be given to the development of combination therapies that add biologically targeted agents to conventional cytotoxic drugs. One example of this type of combination is the addition of the WEE1 inhibitor AZD1775 to the conventional cytotoxic chemotherapeutics, vincristine and irinotecan.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Drug Development/methods , Drug Discovery/methods , Rhabdomyosarcoma , Child , Humans , Research Design
2.
Nat Methods ; 11(8): 821-4, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25042784

ABSTRACT

Cell transplantation into adult zebrafish has lagged behind mouse models owing to the lack of immunocompromised strains. Here we have created rag2(E450fs) mutant zebrafish that have reduced numbers of functional T and B cells but are viable and fecund. Mutant fish engraft muscle, blood stem cells and various cancers. rag2(E450fs) mutant zebrafish are the first immunocompromised zebrafish model that permits robust, long-term engraftment of multiple tissues and cancer.


Subject(s)
Cell Transplantation , DNA-Binding Proteins/genetics , Mutation , Zebrafish/genetics , Aged , Animals , Humans
3.
Proc Natl Acad Sci U S A ; 111(14): 5349-54, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706870

ABSTRACT

Embryonal rhabdomyosarcoma (ERMS) is a common pediatric malignancy of muscle, with relapse being the major clinical challenge. Self-renewing tumor-propagating cells (TPCs) drive cancer relapse and are confined to a molecularly definable subset of ERMS cells. To identify drugs that suppress ERMS self-renewal and induce differentiation of TPCs, a large-scale chemical screen was completed. Glycogen synthase kinase 3 (GSK3) inhibitors were identified as potent suppressors of ERMS growth through inhibiting proliferation and inducing terminal differentiation of TPCs into myosin-expressing cells. In support of GSK3 inhibitors functioning through activation of the canonical WNT/ß-catenin pathway, recombinant WNT3A and stabilized ß-catenin also enhanced terminal differentiation of human ERMS cells. Treatment of ERMS-bearing zebrafish with GSK3 inhibitors activated the WNT/ß-catenin pathway, resulting in suppressed ERMS growth, depleted TPCs, and diminished self-renewal capacity in vivo. Activation of the canonical WNT/ß-catenin pathway also significantly reduced self-renewal of human ERMS, indicating a conserved function for this pathway in modulating ERMS self-renewal. In total, we have identified an unconventional tumor suppressive role for the canonical WNT/ß-catenin pathway in regulating self-renewal of ERMS and revealed therapeutic strategies to target differentiation of TPCs in ERMS.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Rhabdomyosarcoma, Embryonal/pathology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Cell Line , Humans , Rhabdomyosarcoma, Embryonal/enzymology , Rhabdomyosarcoma, Embryonal/metabolism , Zebrafish
4.
Development ; 140(11): 2354-64, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23615277

ABSTRACT

The zebrafish is a powerful genetic model that has only recently been used to dissect developmental pathways involved in oncogenesis. We hypothesized that operative pathways during embryogenesis would also be used for oncogenesis. In an effort to define RAS target genes during embryogenesis, gene expression was evaluated in Tg(hsp70-HRAS(G12V)) zebrafish embryos subjected to heat shock. dusp6 was activated by RAS, and this was used as the basis for a chemical genetic screen to identify small molecules that interfere with RAS signaling during embryogenesis. A KRAS(G12D)-induced zebrafish embryonal rhabdomyosarcoma was then used to assess the therapeutic effects of the small molecules. Two of these inhibitors, PD98059 and TPCK, had anti-tumor activity as single agents in both zebrafish embryonal rhabdomyosarcoma and a human cell line of rhabdomyosarcoma that harbored activated mutations in NRAS. PD98059 inhibited MEK1 whereas TPCK suppressed S6K1 activity; however, the combined treatment completely suppressed eIF4B phosphorylation and decreased translation initiation. Our work demonstrates that the activated pathways in RAS induction during embryogenesis are also important in oncogenesis and that inhibition of these pathways suppresses tumor growth.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Rhabdomyosarcoma/pathology , Signal Transduction , Zebrafish/embryology , ras Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Eukaryotic Initiation Factors/metabolism , Flavonoids/pharmacology , Humans , MAP Kinase Kinase 1/metabolism , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , Ribosomal Protein S6 Kinases/metabolism , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Transgenes , Zebrafish/genetics , Zebrafish Proteins/metabolism
5.
Adv Exp Med Biol ; 916: 219-37, 2016.
Article in English | MEDLINE | ID: mdl-27165356

ABSTRACT

Zebrafish cancer models have greatly advanced our understanding of malignancy in humans. This is made possible due to the unique advantages of the zebrafish model including ex vivo development and large clutch sizes, which enable large-scale genetic and chemical screens. Transparency of the embryo and the creation of adult zebrafish devoid of pigmentation (casper) have permitted unprecedented ability to dynamically visualize cancer progression in live animals. When coupled with fluorescent reporters and transgenic approaches that drive oncogenesis, it is now possible to label entire or subpopulations of cancer cells and follow cancer growth in near real-time. Here, we will highlight aspects of in vivo imaging using the zebrafish and how it has enhanced our understanding of the fundamental aspects of tumor initiation, self-renewal, neovascularization, tumor cell heterogeneity, invasion and metastasis. Importantly, we will highlight the contribution of cancer imaging in zebrafish for drug discovery.


Subject(s)
Diagnostic Imaging , Disease Models, Animal , Neoplasms/pathology , Animals , Animals, Genetically Modified , Disease Progression , Neoplasms/blood supply , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic , Zebrafish/embryology
6.
PLoS Genet ; 9(8): e1003727, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24009521

ABSTRACT

Human cancer genomes are highly complex, making it challenging to identify specific drivers of cancer growth, progression, and tumor maintenance. To bypass this obstacle, we have applied array comparative genomic hybridization (array CGH) to zebrafish embryonal rhabdomyosaroma (ERMS) and utilized cross-species comparison to rapidly identify genomic copy number aberrations and novel candidate oncogenes in human disease. Zebrafish ERMS contain small, focal regions of low-copy amplification. These same regions were commonly amplified in human disease. For example, 16 of 19 chromosomal gains identified in zebrafish ERMS also exhibited focal, low-copy gains in human disease. Genes found in amplified genomic regions were assessed for functional roles in promoting continued tumor growth in human and zebrafish ERMS--identifying critical genes associated with tumor maintenance. Knockdown studies identified important roles for Cyclin D2 (CCND2), Homeobox Protein C6 (HOXC6) and PlexinA1 (PLXNA1) in human ERMS cell proliferation. PLXNA1 knockdown also enhanced differentiation, reduced migration, and altered anchorage-independent growth. By contrast, chemical inhibition of vascular endothelial growth factor (VEGF) signaling reduced angiogenesis and tumor size in ERMS-bearing zebrafish. Importantly, VEGFA expression correlated with poor clinical outcome in patients with ERMS, implicating inhibitors of the VEGF pathway as a promising therapy for improving patient survival. Our results demonstrate the utility of array CGH and cross-species comparisons to identify candidate oncogenes essential for the pathogenesis of human cancer.


Subject(s)
Comparative Genomic Hybridization , Neoplasms/genetics , Oncogenes , Rhabdomyosarcoma, Embryonal/genetics , Zebrafish/genetics , Animals , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , In Situ Hybridization, Fluorescence , Neoplasms/etiology , Oligonucleotide Array Sequence Analysis , Rhabdomyosarcoma, Embryonal/pathology
7.
Mol Cancer Ther ; 22(1): 123-134, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36162055

ABSTRACT

In fusion-negative rhabdomyosarcoma (FN-RMS), a pediatric malignancy with skeletal muscle characteristics, >90% of high-risk patients have mutations that activate the RAS/MEK signaling pathway. We recently discovered that SNAI2, in addition to blocking myogenic differentiation downstream of MEK signaling in FN-RMS, represses proapoptotic BIM expression to protect RMS tumors from ionizing radiation (IR). As clinically relevant concentrations of the MEK inhibitor trametinib elicit poor responses in preclinical xenograft models, we investigated the utility of low-dose trametinib in combination with IR for the treatment of RAS-mutant FN-RMS. We hypothesized that trametinib would sensitize FN-RMS to IR through its downregulation of SNAI2 expression. While we observed little to no difference in myogenic differentiation or cell survival with trametinib treatment alone, robust differentiation and reduced survival were observed after IR. In addition, IR-induced apoptosis was significantly increased in FN-RMS cells treated concurrently with trametinib, as was increased BIM expression. SNAI2's role in these processes was established using overexpression rescue experiments, where overexpression of SNAI2 prevented IR-induced myogenic differentiation and apoptosis. Moreover, combining MEK inhibitor with IR resulted in complete tumor regression and a 2- to 4-week delay in event-free survival (EFS) in preclinical xenograft and patient-derived xenograft models. Our findings demonstrate that the combination of MEK inhibition and IR results in robust differentiation and apoptosis, due to the reduction of SNAI2, which leads to extended EFS in FN-RMS. SNAI2 thus is a potential biomarker of IR insensitivity and target for future therapies to sensitize aggressive sarcomas to IR.


Subject(s)
Rhabdomyosarcoma , Child , Humans , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/radiotherapy , Cell Differentiation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Mitogen-Activated Protein Kinase Kinases , Cell Line, Tumor , Snail Family Transcription Factors
8.
Mol Cell Biol ; 43(11): 547-565, 2023.
Article in English | MEDLINE | ID: mdl-37882064

ABSTRACT

Rhabdomyosarcoma (RMS) is a pediatric malignancy of the muscle with characteristics of cells blocked in differentiation. NOTCH1 is an oncogene that promotes self-renewal and blocks differentiation in the fusion negative-RMS sub-type. However, how NOTCH1 expression is transcriptionally maintained in tumors is unknown. Analyses of SNAI2 and CTCF chromatin binding and HiC analyses revealed a conserved SNAI2/CTCF overlapping peak downstream of the NOTCH1 locus marking a sub-topologically associating domain (TAD) boundary. Deletion of the SNAI2-CTCF peak showed that it is essential for NOTCH1 expression and viability of FN-RMS cells. Reintroducing constitutively activated NOTCH1-ΔE in cells with the SNAI2-CTCF peak deleted restored cell-viability. Ablation of SNAI2 using CRISPR/Cas9 reagents resulted in the loss of majority of RD and SMS-CTR FN-RMS cells. However, the few surviving clones that repopulate cultures have recovered NOTCH1. Cells that re-establish NOTCH1 expression after SNAI2 ablation are unable to differentiate robustly as SNAI2 shRNA knockdown cells; yet, SNAI2-ablated cells continued to be exquisitely sensitive to ionizing radiation. Thus, we have uncovered a novel mechanism by which SNAI2 and CTCF maintenance of a sub-TAD boundary promotes rather than represses NOTCH1 expression. Further, we demonstrate that SNAI2 suppression of apoptosis post-radiation is independent of SNAI2/NOTCH1 effects on self-renewal and differentiation.


Subject(s)
Chromatin , Rhabdomyosarcoma , Child , Humans , CCCTC-Binding Factor/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Rhabdomyosarcoma/genetics , RNA, Small Interfering/genetics , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
9.
Elife ; 122023 06 02.
Article in English | MEDLINE | ID: mdl-37266578

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.


Subject(s)
Cerebellar Neoplasms , Rhabdomyosarcoma, Embryonal , Animals , Carcinogenesis , Mutation , Proto-Oncogene Proteins p21(ras)/metabolism , Rhabdomyosarcoma, Embryonal/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/genetics , Zebrafish/metabolism
10.
Blood ; 115(16): 3296-303, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20056790

ABSTRACT

Self-renewal is a feature of cancer and can be assessed by cell transplantation into immune-compromised or immune-matched animals. However, studies in zebrafish have been severely limited by lack of these reagents. Here, Myc-induced T-cell acute lymphoblastic leukemias (T-ALLs) have been made in syngeneic, clonal zebrafish and can be transplanted into sibling animals without the need for immune suppression. These studies show that self-renewing cells are abundant in T-ALL and comprise 0.1% to 15.9% of the T-ALL mass. Large-scale single-cell transplantation experiments established that T-ALLs can be initiated from a single cell and that leukemias exhibit wide differences in tumor-initiating potential. T-ALLs also can be introduced into clonal-outcrossed animals, and T-ALLs arising in mixed genetic backgrounds can be transplanted into clonal recipients without the need for major histocompatibility complex matching. Finally, high-throughput imaging methods are described that allow large numbers of fluorescent transgenic animals to be imaged simultaneously, facilitating the rapid screening of engrafted animals. Our experiments highlight the large numbers of zebrafish that can be experimentally assessed by cell transplantation and establish new high-throughput methods to functionally interrogate gene pathways involved in cancer self-renewal.


Subject(s)
Disease Models, Animal , Neoplasm Transplantation/methods , Neoplastic Stem Cells/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cell Separation , Flow Cytometry , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics
11.
Mol Cancer Ther ; 21(1): 170-183, 2022 01.
Article in English | MEDLINE | ID: mdl-34737198

ABSTRACT

Oncogenic RAS signaling is an attractive target for fusion-negative rhabdomyosarcoma (FN-RMS). Our study validates the role of the ERK MAPK effector pathway in mediating RAS dependency in a panel of H/NRASQ61X mutant RMS cells and correlates in vivo efficacy of the MEK inhibitor trametinib with pharmacodynamics of ERK activity. A screen is used to identify trametinib-sensitizing targets, and combinations are evaluated in cells and tumor xenografts. We find that the ERK MAPK pathway is central to H/NRASQ61X dependency in RMS cells; however, there is poor in vivo response to clinically relevant exposures with trametinib, which correlates with inefficient suppression of ERK activity. CRISPR screening points to vertical inhibition of the RAF-MEK-ERK cascade by cosuppression of MEK and either CRAF or ERK. CRAF is central to rebound pathway activation following MEK or ERK inhibition. Concurrent CRAF suppression and MEK or ERK inhibition, or concurrent pan-RAF and MEK/ERK inhibition (pan-RAFi + MEKi/ERKi), or concurrent MEK and ERK inhibition (MEKi + ERKi) all synergistically block ERK activity and induce myogenic differentiation and apoptosis. In vivo assessment of pan-RAFi + ERKi or MEKi + ERKi potently suppress growth of H/NRASQ61X RMS tumor xenografts, with pan-RAFi + ERKi being more effective and better tolerated. We conclude that CRAF reactivation limits the activity of single-agent MEK/ERK inhibitors in FN-RMS. Vertical targeting of the RAF-MEK-ERK cascade and particularly cotargeting of CRAF and MEK or ERK, or the combination of pan-RAF inhibitors with MEK or ERK inhibitors, have synergistic activity and potently suppress H/NRASQ61X mutant RMS tumor growth.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/genetics , Proto-Oncogene Proteins B-raf/metabolism , Rhabdomyosarcoma/genetics , Animals , Apoptosis , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Female , Genes, ras , Humans , Mice , Rhabdomyosarcoma/pathology , Transfection
12.
Zebrafish ; 18(4): 293-296, 2021 08.
Article in English | MEDLINE | ID: mdl-34030492

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.


Subject(s)
Hemangiosarcoma , Neoplasm Transplantation , Zebrafish , Animals , Disease Models, Animal , Hemangiosarcoma/pathology , Suspensions , Tumor Microenvironment
13.
Nat Commun ; 12(1): 192, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420019

ABSTRACT

Rhabdomyosarcoma (RMS) is an aggressive pediatric malignancy of the muscle, that includes Fusion Positive (FP)-RMS harboring PAX3/7-FOXO1 and Fusion Negative (FN)-RMS commonly with RAS pathway mutations. RMS express myogenic master transcription factors MYOD and MYOG yet are unable to terminally differentiate. Here, we report that SNAI2 is highly expressed in FN-RMS, is oncogenic, blocks myogenic differentiation, and promotes growth. MYOD activates SNAI2 transcription via super enhancers with striped 3D contact architecture. Genome wide chromatin binding analysis demonstrates that SNAI2 preferentially binds enhancer elements and competes with MYOD at a subset of myogenic enhancers required for terminal differentiation. SNAI2 also suppresses expression of a muscle differentiation program modulated by MYOG, MEF2, and CDKN1A. Further, RAS/MEK-signaling modulates SNAI2 levels and binding to chromatin, suggesting that the differentiation blockade by oncogenic RAS is mediated in part by SNAI2. Thus, an interplay between SNAI2, MYOD, and RAS prevents myogenic differentiation and promotes tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Cell Differentiation , MyoD Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , Snail Family Transcription Factors/metabolism , Animals , Carcinogenesis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , MEF2 Transcription Factors/metabolism , Male , Mice , Mice, SCID , Muscle Development/genetics , MyoD Protein/genetics , Myogenin/metabolism , Oncogene Proteins, Fusion/genetics , Oncogenes , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Embryonal/genetics , Snail Family Transcription Factors/genetics , Transcriptome
14.
Cancer Res ; 81(21): 5451-5463, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34462275

ABSTRACT

Ionizing radiation (IR) and chemotherapy are mainstays of treatment for patients with rhabdomyosarcoma, yet the molecular mechanisms that underlie the success or failure of radiotherapy remain unclear. The transcriptional repressor SNAI2 was previously identified as a key regulator of IR sensitivity in normal and malignant stem cells through its repression of the proapoptotic BH3-only gene PUMA/BBC3. Here, we demonstrate a clear correlation between SNAI2 expression levels and radiosensitivity across multiple rhabdomyosarcoma cell lines. Modulating SNAI2 levels in rhabdomyosarcoma cells through its overexpression or knockdown altered radiosensitivity in vitro and in vivo. SNAI2 expression reliably promoted overall cell growth and inhibited mitochondrial apoptosis following exposure to IR, with either variable or minimal effects on differentiation and senescence, respectively. Importantly, SNAI2 knockdown increased expression of the proapoptotic BH3-only gene BIM, and chromatin immunoprecipitation sequencing experiments established that SNAI2 is a direct repressor of BIM/BCL2L11. Because the p53 pathway is nonfunctional in the rhabdomyosarcoma cells used in this study, we have identified a new, p53-independent SNAI2/BIM signaling axis that could potentially predict clinical responses to IR treatment and be exploited to improve rhabdomyosarcoma therapy. SIGNIFICANCE: SNAI2 is identified as a major regulator of radiation-induced apoptosis in rhabdomyosarcoma through previously unknown mechanisms independent of p53.


Subject(s)
Bcl-2-Like Protein 11/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic/radiation effects , Radiation, Ionizing , Rhabdomyosarcoma/prevention & control , Snail Family Transcription Factors/metabolism , Animals , Apoptosis , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Biomarkers, Tumor/genetics , Cell Cycle , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, SCID , RNA-Seq , Rhabdomyosarcoma/etiology , Rhabdomyosarcoma/pathology , Snail Family Transcription Factors/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Dev Biol ; 313(2): 568-83, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18068699

ABSTRACT

Neural crest-derived pigment cell development has been used extensively to study cell fate specification, migration, proliferation, survival and differentiation. Many of the genes and regulatory mechanisms required for pigment cell development are conserved across vertebrates. The zebrafish mutant colgate (col)/histone deacetylase1 (hdac1) has reduced numbers, delayed differentiation and decreased migration of neural crest-derived melanophores and their precursors. In hdac1(col) mutants normal numbers of premigratory neural crest cells are induced. Later, while there is only a slight reduction in the number of neural crest cells in hdac1(col) mutants, there is a severe reduction in the number of mitfa-positive melanoblasts suggesting that hdac1 is required for melanoblast specification. Concomitantly, there is a significant increase in and prolonged expression of foxd3 in neural crest cells in hdac1(col) mutants. We found that partially reducing Foxd3 expression in hdac1(col) mutants rescues mitfa expression and the melanophore defects in hdac1(col) mutants. Furthermore, we demonstrate the ability of Foxd3 to physically interact at the mitfa promoter. Because mitfa is required for melanoblast specification and development, our results suggest that hdac1 is normally required to suppress neural crest foxd3 expression thus de-repressing mitfa resulting in melanogenesis by a subset of neural crest-derived cells.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases/physiology , Microphthalmia-Associated Transcription Factor/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , Zebrafish/physiology , Animals , Base Sequence , Binding Sites , Cell Movement , Electrophoretic Mobility Shift Assay , Embryo, Nonmammalian , Histone Deacetylase 1 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , In Situ Hybridization , Melanophores/cytology , Melanophores/metabolism , Melanophores/physiology , Microinjections , Models, Biological , Molecular Sequence Data , Mutation , Neural Crest/cytology , Neural Crest/embryology , Oligonucleotides, Antisense/pharmacology , Promoter Regions, Genetic , Protein Binding , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
16.
Mech Dev ; 124(9-10): 682-98, 2007.
Article in English | MEDLINE | ID: mdl-17716875

ABSTRACT

Vertebrate gastrulation involves the coordinated movements of populations of cells. These movements include cellular rearrangements in which cells polarize along their medio-lateral axes leading to cell intercalations that result in elongation of the body axis. Molecular analysis of this process has implicated the non-canonical Wnt/Frizzled signaling pathway that is similar to the planar cell polarity pathway (PCP) in Drosophila. Here we describe a zebrafish mutant, colgate (col), which displays defects in the extension of the body axis and the migration of branchiomotor neurons. Activation of the non-canonical Wnt/PCP pathway in these mutant embryos by overexpressing DeltaNdishevelled, rho kinase2 and van gogh-like protein 2 (vangl2) rescues the extension defects suggesting that col acts as a positive regulator of the non-canonical Wnt/PCP pathway. Further, we show that col normally regulates the caudal migration of nVII facial hindbrain branchiomotor neurons and that the mutant phenotype can be rescued by misexpression of vangl2 independent of the Wnt/PCP pathway. We cloned the col locus and found that it encodes histone deacetylase1 (hdac1). Our previous results and studies by others have implicated hdac1 in repressing the canonical Wnt pathway. Here, we demonstrate novel roles for zebrafish hdac1 in activating non-canonical Wnt/PCP signaling underlying axial extension and in promoting Wnt-independent caudal migration of a subset of hindbrain branchiomotor neurons.


Subject(s)
Axons/physiology , Body Patterning/physiology , Cell Movement/physiology , Histone Deacetylases/physiology , Motor Neurons/physiology , Signal Transduction/physiology , Wnt Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Axons/enzymology , Body Patterning/genetics , Cell Movement/genetics , Cell Polarity/genetics , Cell Polarity/physiology , Histone Deacetylase 1 , Histone Deacetylases/genetics , Mutation , Rhombencephalon/cytology , Rhombencephalon/embryology , Rhombencephalon/enzymology , Signal Transduction/genetics , Wnt Proteins/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
17.
Elife ; 72018 09 07.
Article in English | MEDLINE | ID: mdl-30192230

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.


Subject(s)
Rhabdomyosarcoma, Embryonal/metabolism , Rhabdomyosarcoma, Embryonal/pathology , Tumor Suppressor Protein p53/deficiency , Zebrafish/metabolism , Animals , Cell Count , Gene Deletion , Gene Expression Regulation, Neoplastic , Hemangiosarcoma/metabolism , Hemangiosarcoma/pathology , Homozygote , Leukemia/metabolism , Leukemia/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Rhabdomyosarcoma, Embryonal/genetics , Survival Analysis , Tumor Suppressor Protein p53/metabolism , Zebrafish/genetics
18.
Cell Stem Cell ; 22(3): 414-427.e6, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29499154

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.


Subject(s)
Cell Self Renewal , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Neoplastic Stem Cells/pathology , Rhabdomyosarcoma/pathology , Signal Transduction , Zebrafish Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplastic Stem Cells/metabolism , Rhabdomyosarcoma/genetics , Xenograft Model Antitumor Assays
19.
Elife ; 62017 01 12.
Article in English | MEDLINE | ID: mdl-28080960

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.


Subject(s)
MyoD Protein/metabolism , Myogenic Regulatory Factor 5/metabolism , Rhabdomyosarcoma/physiopathology , Transcription, Genetic , Animals , Animals, Genetically Modified , Chromatin Immunoprecipitation , Humans , Sequence Analysis, DNA , Zebrafish
20.
Cell Rep ; 19(11): 2304-2318, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28614716

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.


Subject(s)
MEF2 Transcription Factors/metabolism , Receptor, Notch1/metabolism , Rhabdomyosarcoma, Embryonal/metabolism , Snail Family Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Humans , Rhabdomyosarcoma, Embryonal/pathology , Signal Transduction , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL