Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Cell Rep Med ; 5(3): 101472, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38508140

ABSTRACT

Anaplastic large cell lymphoma (ALCL) is an aggressive, CD30+ T cell lymphoma of children and adults. ALK fusion transcripts or mutations in the JAK-STAT pathway are observed in most ALCL tumors, but the mechanisms underlying tumorigenesis are not fully understood. Here, we show that dysregulated STAT3 in ALCL cooccupies enhancers with master transcription factors BATF3, IRF4, and IKZF1 to form a core regulatory circuit that establishes and maintains the malignant cell state in ALCL. Critical downstream targets of this network in ALCL cells include the protooncogene MYC, which requires active STAT3 to facilitate high levels of MYC transcription. The core autoregulatory transcriptional circuitry activity is reinforced by MYC binding to the enhancer regions associated with STAT3 and each of the core regulatory transcription factors. Thus, activation of STAT3 provides the crucial link between aberrant tyrosine kinase signaling and the core transcriptional machinery that drives tumorigenesis and creates therapeutic vulnerabilities in ALCL.


Subject(s)
Lymphoma, Large-Cell, Anaplastic , Signal Transduction , Adult , Child , Humans , Signal Transduction/genetics , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/metabolism , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Cell Transformation, Neoplastic , Carcinogenesis/genetics , STAT3 Transcription Factor/genetics
2.
Article in English | MEDLINE | ID: mdl-37937078

ABSTRACT

Introduction: Myelodysplastic syndrome (MDS) is a heterogeneous group of clonal hematopoietic disorders characterized by ineffective hematopoiesis, cytopenias, and dysplasia. The gene encoding ten-eleven translocation 2 (tet2), a dioxygenase enzyme that catalyzes the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine, is a recurrently mutated tumor suppressor gene in MDS and other myeloid malignancies. Previously, we reported a stable zebrafish line with a loss-of-function mutation in the tet2 gene. The tet2m/m-mutant zebrafish developed a pre-MDS state with kidney marrow dysplasia, but normal circulating blood counts by 11 months of age and accompanying anemia, signifying the onset of MDS, by 24 months of age. Methods: In the current study, we collected progenitor cells from the kidney marrows of the adult tet2m/m and tet2wt/wt fish at 4 and 15 months of age and conducted enhanced reduced representation of bisulfite sequencing (ERRBS) and bulk RNA-seq to measure changes in DNA methylation and gene expression of hematopoietic stem and progenitor cells (HSPCs). Results and discussion: A global increase in DNA methylation of gene promoter regions and CpG islands was observed in tet2m/m HSPCs at 4 months of age when compared with the wild type. Furthermore, hypermethylated genes were significantly enriched for targets of SUZ12 and the metal-response-element-binding transcription factor 2 (MTF2)-involved in the polycomb repressive complex 2 (PRC2). However, between 4 and 15 months of age, we observed a paradoxical global decrease in DNA methylation in tet2m/m HSPCs. Gene expression analyses identified upregulation of genes associated with mTORC1 signaling and interferon gamma and alpha responses in tet2m/m HSPCs at 4 months of age when compared with the wild type. Downregulated genes in HSPCs of tet2-mutant fish at 4 months of age were enriched for cell cycle regulation, heme metabolism, and interleukin 2 (IL2)/signal transducer and activator of transcription 5 (STAT5) signaling, possibly related to increased self-renewal and clonal advantage in HSPCs with tet2 loss of function. Finally, there was an overall inverse correlation between overall increased promoter methylation and gene expression.

3.
J Endod ; 49(7): 836-845, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37182792

ABSTRACT

INTRODUCTION: Concentrated growth factor (CGF) is the third-generation platelet concentrate product. This study aimed to evaluate whether the use of CGF during endodontic microsurgery had a positive influence on surgical outcomes. METHODS: Fifty-four patients who underwent endodontic microsurgery from January 2017 to November 2021 were enrolled. They were assigned to the CGF and the control groups according to whether CGF was used during the surgery and followed up at 6, 12, and 18 months after surgery. Preoperative classification of the cases and follow-up radiographic outcomes were based on Kim's classification and Molven's criteria, respectively, and evaluated by 2 calibrated endodontists. The Student t test and χ2 test were used to assess the baseline of 2 groups. Rank sum test was used to determine whether CGF had an impact on the surgical outcome. RESULTS: Thirty-one patients (41 periapical lesion sites) were included in the CGF group, and 23 patients (26 periapical lesion sites) were included in the control group. The overall success rate of endodontic microsurgery was greater than 90%. The baseline of the 2 groups had no difference (P < .05). In the CGF group, the success rate was always 100% in 3 follow-ups, whereas the success rate was 84.2%, 92.8%, and 90%, respectively, in the control group. The success rate between the CGF group and the control group was statistically significant in all 3 follow-up points (P < .05). CONCLUSIONS: The application of CGF during endodontic microsurgery might have a positive influence on surgical outcomes, thus, its prognosis. However, higher-grade evidence is needed to demonstrate its role.


Subject(s)
Intercellular Signaling Peptides and Proteins , Microsurgery , Humans , Treatment Outcome , Cross-Sectional Studies , Prognosis , Intercellular Signaling Peptides and Proteins/therapeutic use
4.
Br J Haematol ; 201(3): 489-501, 2023 05.
Article in English | MEDLINE | ID: mdl-36746437

ABSTRACT

TET2 inactivating mutations serve as initiating genetic lesions in the transformation of haematopoietic stem and progenitor cells (HSPCs). In this study, we analysed known drugs in zebrafish embryos for their ability to selectively kill tet2-mutant HSPCs in vivo. We found that the exportin 1 (XPO1) inhibitors, selinexor and eltanexor, selectively kill tet2-mutant HSPCs. In serial replating colony assays, these small molecules were selectively active in killing murine Tet2-deficient Lineage-, Sca1+, Kit+ (LSK) cells, and also TET2-inactivated human acute myeloid leukaemia (AML) cells. Selective killing of TET2-mutant HSPCs and human AML cells by these inhibitors was due to increased levels of apoptosis, without evidence of DNA damage based on increased γH2AX expression. The finding that TET2 loss renders HSPCs and AML cells selectively susceptible to cell death induced by XPO1 inhibitors provides preclinical evidence of the selective activity of these drugs, justifying further clinical studies of these small molecules for the treatment of TET2-mutant haematopoietic malignancies, and to suppress clonal expansion in age-related TET2-mutant clonal haematopoiesis.


Subject(s)
Dioxygenases , Leukemia, Myeloid, Acute , Animals , Humans , Mice , Zebrafish , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , DNA-Binding Proteins/genetics , Dioxygenases/metabolism , Exportin 1 Protein
6.
Sci Adv ; 7(43): eabe0834, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34669465

ABSTRACT

Neuroblastoma cell identity depends on a core regulatory circuit (CRC) of transcription factors that collaborate with MYCN to drive the oncogenic gene expression program. For neuroblastomas dependent on the adrenergic CRC, treatment with retinoids can inhibit cell growth and induce differentiation. Here, we show that when MYCN-amplified neuroblastoma cells are treated with retinoic acid, histone H3K27 acetylation and methylation become redistributed to decommission super-enhancers driving the expression of PHOX2B and GATA3, together with the activation of new super-enhancers that drive high levels of MEIS1 and SOX4 expression. These findings indicate that treatment with retinoids can reprogram the enhancer landscape, resulting in down-regulation of MYCN expression, while establishing a new retino-sympathetic CRC that causes proliferative arrest and sympathetic differentiation. Thus, we provide mechanisms that account for the beneficial effects of retinoids in high-risk neuroblastoma and explain the rapid down-regulation of expression of MYCN despite massive levels of amplification of this gene.

7.
Oncogene ; 40(38): 5718-5729, 2021 09.
Article in English | MEDLINE | ID: mdl-34331013

ABSTRACT

Melanomas driven by loss of the NF1 tumor suppressor have a high risk of treatment failure and effective therapies have not been developed. Here we show that loss-of-function mutations of nf1 and pten result in aggressive melanomas in zebrafish, representing the first animal model of NF1-mutant melanomas harboring PTEN loss. MEK or PI3K inhibitors show little activity when given alone due to cross-talk between the pathways, and high toxicity when given together. The mTOR inhibitors, sirolimus, everolimus, and temsirolimus, were the most active single agents tested, potently induced tumor-suppressive autophagy, but not apoptosis. Because addition of the BCL2 inhibitor venetoclax resulted in compensatory upregulation of MCL1, we established a three-drug combination composed of sirolimus, venetoclax, and the MCL1 inhibitor S63845. This well-tolerated drug combination potently and synergistically induces apoptosis in both zebrafish and human NF1/PTEN-deficient melanoma cells, providing preclinical evidence justifying an early-stage clinical trial in patients with NF1/PTEN-deficient melanoma.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , MTOR Inhibitors/administration & dosage , Melanoma/drug therapy , Neurofibromin 1/genetics , PTEN Phosphohydrolase/genetics , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Thiophenes/administration & dosage , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Everolimus/administration & dosage , Everolimus/pharmacology , Humans , Loss of Function Mutation , MTOR Inhibitors/pharmacology , Melanoma/genetics , Melanoma/pathology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrimidines/pharmacology , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Xenograft Model Antitumor Assays , Zebrafish
8.
Nat Chem Biol ; 17(9): 954-963, 2021 09.
Article in English | MEDLINE | ID: mdl-33972797

ABSTRACT

The peptidyl-prolyl isomerase, Pin1, is exploited in cancer to activate oncogenes and inactivate tumor suppressors. However, despite considerable efforts, Pin1 has remained an elusive drug target. Here, we screened an electrophilic fragment library to identify covalent inhibitors targeting Pin1's active site Cys113, leading to the development of Sulfopin, a nanomolar Pin1 inhibitor. Sulfopin is highly selective, as validated by two independent chemoproteomics methods, achieves potent cellular and in vivo target engagement and phenocopies Pin1 genetic knockout. Pin1 inhibition had only a modest effect on cancer cell line viability. Nevertheless, Sulfopin induced downregulation of c-Myc target genes, reduced tumor progression and conferred survival benefit in murine and zebrafish models of MYCN-driven neuroblastoma, and in a murine model of pancreatic cancer. Our results demonstrate that Sulfopin is a chemical probe suitable for assessment of Pin1-dependent pharmacology in cells and in vivo, and that Pin1 warrants further investigation as a potential cancer drug target.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-myc/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
9.
Cancer Res ; 81(11): 2995-3007, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33602789

ABSTRACT

One of the greatest barriers to curative treatment of neuroblastoma is its frequent metastatic outgrowth prior to diagnosis, especially in cases driven by amplification of the MYCN oncogene. However, only a limited number of regulatory proteins that contribute to this complex MYCN-mediated process have been elucidated. Here we show that the growth arrest-specific 7 (GAS7) gene, located at chromosome band 17p13.1, is preferentially deleted in high-risk MYCN-driven neuroblastoma. GAS7 expression was also suppressed in MYCN-amplified neuroblastoma lacking 17p deletion. GAS7 deficiency led to accelerated metastasis in both zebrafish and mammalian models of neuroblastoma with overexpression or amplification of MYCN. Analysis of expression profiles and the ultrastructure of zebrafish neuroblastoma tumors with MYCN overexpression identified that GAS7 deficiency led to (i) downregulation of genes involved in cell-cell interaction, (ii) loss of contact among tumor cells as critical determinants of accelerated metastasis, and (iii) increased levels of MYCN protein. These results provide the first genetic evidence that GAS7 depletion is a critical early step in the cascade of events culminating in neuroblastoma metastasis in the context of MYCN overexpression. SIGNIFICANCE: Heterozygous deletion or MYCN-mediated repression of GAS7 in neuroblastoma releases an important brake on tumor cell dispersion and migration to distant sites, providing a novel mechanism underlying tumor metastasis in MYCN-driven neuroblastoma.See related commentary by Menard, p. 2815.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Marrow Neoplasms/secondary , Chromosome Deletion , Gene Expression Regulation, Neoplastic , N-Myc Proto-Oncogene Protein/metabolism , Nerve Tissue Proteins/deficiency , Neuroblastoma/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Bone Marrow Neoplasms/genetics , Bone Marrow Neoplasms/metabolism , Cell Proliferation , Humans , Mice , Mice, SCID , N-Myc Proto-Oncogene Protein/genetics , Nerve Tissue Proteins/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zebrafish
10.
Dis Model Mech ; 13(8)2020 08 27.
Article in English | MEDLINE | ID: mdl-32651197

ABSTRACT

Polycomb repressive complex 2 (PRC2) is an epigenetic regulator of gene expression that possesses histone methyltransferase activity. PRC2 trimethylates lysine 27 of histone H3 proteins (H3K27me3) as a chromatin modification associated with repressed transcription of genes frequently involved in cell proliferation or self-renewal. Loss-of-function mutations in the PRC2 core subunit SUZ12 have been identified in a variety of tumors, including malignant peripheral nerve sheath tumors (MPNSTs). To determine the consequences of SUZ12 loss in the pathogenesis of MPNST and other cancers, we used CRISPR-Cas9 to disrupt the open reading frame of each of two orthologous suz12 genes in zebrafish: suz12a and suz12b We generated these knockout alleles in the germline of our previously described p53 (also known as tp53)- and nf1-deficient zebrafish model of MPNSTs. Loss of suz12 significantly accelerated the onset and increased the penetrance of MPNSTs compared to that in control zebrafish. Moreover, in suz12-deficient zebrafish, we detected additional types of tumors besides MPNSTs, including leukemia with histological characteristics of lymphoid malignancies, soft tissue sarcoma and pancreatic adenocarcinoma, which were not detected in p53/nf1-deficient control fish, and are also contained in the human spectrum of SUZ12-deficient malignancies identified in the AACR Genie database. The suz12-knockout tumors displayed reduced or abolished H3K27me3 epigenetic marks and upregulation of gene sets reported to be targeted by PRC2. Thus, these zebrafish lines with inactivation of suz12 in combination with loss of p53/nf1 provide a model of human MPNSTs and multiple other tumor types, which will be useful for mechanistic studies of molecular pathogenesis and targeted therapy with small molecule inhibitors.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Silencing , Neurofibromin 1/genetics , Neurofibrosarcoma/genetics , Tumor Suppressor Protein p53/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Neurofibromin 1/deficiency , Neurofibrosarcoma/drug therapy , Neurofibrosarcoma/metabolism , Neurofibrosarcoma/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Sarcoma/genetics , Sarcoma/metabolism , Sarcoma/pathology , Signal Transduction , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/pathology , Tumor Suppressor Protein p53/deficiency , Zebrafish/metabolism , Zebrafish Proteins/deficiency
11.
Leukemia ; 34(11): 2992-3006, 2020 11.
Article in English | MEDLINE | ID: mdl-32572188

ABSTRACT

Inactivating mutations in TET2 serve as an initiating genetic lesion in the transformation of hematopoietic stem and progenitor cells (HSPCs). Thus, effective therapy for this subset of patients would ideally include drugs that are selectively lethal in TET2-mutant HSPCs, at dosages that spare normal HSPCs. In this study, we tested 129 FDA-approved anticancer drugs in a tet2-deficient zebrafish model and showed that topoisomerase 1 (TOP1)-targeted drugs and PARP1 inhibitors selectively kill tet2-mutant HSPCs. We found that Tet2-deficient murine bone marrow progenitors and CRISPR-Cas9-induced TET2-mutant human AML cells were more sensitive to both classes of drugs compared with matched control cells. The mechanism underlying the selective killing of TET2-mutant blood cells by these drugs was due to aberrantly low levels of tyrosyl-DNA phosphodiesterase 1 (TDP1), an enzyme that is important for removing TOP1 cleavage complexes (TOP1cc). Low TDP1 levels yield sensitivity to TOP1-targeted drugs or PARP1 inhibitors and an inability to remove TOP1 cleavage complexes, leading to DNA double-strand breaks and cell death. The finding that TET2 mutations render HSPCs uniquely vulnerable to disruption of TOP1 and PARP1 activity may therefore represent a unique opportunity to use relatively low dosages of these drugs for the "precision therapy" of TET2-mutant myeloid malignancies.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Synthetic Lethal Mutations , Topoisomerase I Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Cell Survival/genetics , DNA Breaks, Double-Stranded/drug effects , Dioxygenases , Genotype , Humans , Mice , Mice, Knockout , Phenotype , Topotecan/pharmacology , Zebrafish
12.
Cell ; 181(3): 702-715.e20, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32315619

ABSTRACT

Protein phosphatase 2A (PP2A) enzymes can suppress tumors, but they are often inactivated in human cancers overexpressing inhibitory proteins. Here, we identify a class of small-molecule iHAPs (improved heterocyclic activators of PP2A) that kill leukemia cells by allosterically assembling a specific heterotrimeric PP2A holoenzyme consisting of PPP2R1A (scaffold), PPP2R5E (B56ε, regulatory), and PPP2CA (catalytic) subunits. One compound, iHAP1, activates this complex but does not inhibit dopamine receptor D2, a mediator of neurologic toxicity induced by perphenazine and related neuroleptics. The PP2A complex activated by iHAP1 dephosphorylates the MYBL2 transcription factor on Ser241, causing irreversible arrest of leukemia and other cancer cells in prometaphase. In contrast, SMAPs, a separate class of compounds, activate PP2A holoenzymes containing a different regulatory subunit, do not dephosphorylate MYBL2, and arrest tumor cells in G1 phase. Our findings demonstrate that small molecules can serve as allosteric switches to activate distinct PP2A complexes with unique substrates.


Subject(s)
Protein Phosphatase 2/metabolism , Apoptosis , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Enzyme Activators/metabolism , G1 Phase , Humans , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , Phenothiazines/pharmacology , Phosphorylation , Protein Phosphatase 2/physiology , Protein Subunits/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism
13.
Nat Chem Biol ; 16(6): 635-643, 2020 06.
Article in English | MEDLINE | ID: mdl-32251410

ABSTRACT

Doublecortin like kinase 1 (DCLK1) is an understudied kinase that is upregulated in a wide range of cancers, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about its potential as a therapeutic target. We used chemoproteomic profiling and structure-based design to develop a selective, in vivo-compatible chemical probe of the DCLK1 kinase domain, DCLK1-IN-1. We demonstrate activity of DCLK1-IN-1 against clinically relevant patient-derived PDAC organoid models and use a combination of RNA-sequencing, proteomics and phosphoproteomics analysis to reveal that DCLK1 inhibition modulates proteins and pathways associated with cell motility in this context. DCLK1-IN-1 will serve as a versatile tool to investigate DCLK1 biology and establish its role in cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Movement , Doublecortin Protein , Doublecortin-Like Kinases , Drug Screening Assays, Antitumor , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors/pharmacokinetics , Proteomics , Rats , Structure-Activity Relationship , Zebrafish , Pancreatic Neoplasms
14.
Zebrafish ; 16(4): 421-426, 2019 08.
Article in English | MEDLINE | ID: mdl-31063042

ABSTRACT

In July 2018, the 11th Zebrafish Disease Models Conference (ZDM11) was held at Leiden University, The Netherlands, providing an excellent international opportunity for scientific presentations and collaborative discussion regarding the modeling of disease using zebrafish. Much like the original ZDM1, which was also hosted in Leiden in 2007, immunology and cancer had a strong presence at ZDM11, with zebrafish still proving an invaluable tool to interrogate their disease genetics and progression in vivo. In addition, ZDM11 built upon the inclusion and development of other key areas making use of zebrafish disease models, with sessions on neuroscience, behavior, muscle, skeletal and cardiac disease, and more. ZDM11 also highlighted the rapid progression and application of new and exciting technologies to assist in the generation and analysis of zebrafish disease models, including Crispr/Cas9 gene targeting tools, electroporation techniques, computational analysis, drug screening pipelines, and advances in vivo imaging such as high-resolution correlative electron microscopy and lightsheet microscopy. Here, we provide a summary of the ZDM11 conference proceedings, giving an overview of the stimulating science presented across 4 days and 13 conference sessions.


Subject(s)
Fish Diseases , Zebrafish , Animals , Congresses as Topic , Disease Models, Animal , Fish Diseases/etiology , Fish Diseases/immunology , Neoplasms/etiology , Netherlands
15.
PLoS Genet ; 15(4): e1008039, 2019 04.
Article in English | MEDLINE | ID: mdl-30970016

ABSTRACT

The SWI/SNF-family chromatin remodeling protein ATRX is a tumor suppressor in sarcomas, gliomas and other malignancies. Its loss of function facilitates the alternative lengthening of telomeres (ALT) pathway in tumor cells, while it also affects Polycomb repressive complex 2 (PRC2) silencing of its target genes. To further define the role of inactivating ATRX mutations in carcinogenesis, we knocked out atrx in our previously reported p53/nf1-deficient zebrafish line that develops malignant peripheral nerve sheath tumors and gliomas. Complete inactivation of atrx using CRISPR/Cas9 was lethal in developing fish and resulted in an alpha-thalassemia-like phenotype including reduced alpha-globin expression. In p53/nf1-deficient zebrafish neither peripheral nerve sheath tumors nor gliomas showed accelerated onset in atrx+/- fish, but these fish developed various tumors that were not observed in their atrx+/+ siblings, including epithelioid sarcoma, angiosarcoma, undifferentiated pleomorphic sarcoma and rare types of carcinoma. These cancer types are included in the AACR Genie database of human tumors associated with mutant ATRX, indicating that our zebrafish model reliably mimics a role for ATRX-loss in the early pathogenesis of these human cancer types. RNA-seq of p53/nf1- and p53/nf1/atrx-deficient tumors revealed that down-regulation of telomerase accompanied ALT-mediated lengthening of the telomeres in atrx-mutant samples. Moreover, inactivating mutations in atrx disturbed PRC2-target gene silencing, indicating a connection between ATRX loss and PRC2 dysfunction in cancer development.


Subject(s)
Sarcoma, Experimental/etiology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , X-linked Nuclear Protein/deficiency , X-linked Nuclear Protein/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Carcinogenesis/genetics , Carcinogenesis/metabolism , Disease Models, Animal , Erythropoiesis , Female , Gene Knockout Techniques , Globins/genetics , Humans , Loss of Function Mutation , Male , Neurofibromin 1/deficiency , Neurofibromin 1/genetics , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Telomere Homeostasis/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
16.
Cancer Res ; 79(9): 2136-2151, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30862716

ABSTRACT

Alterations in lipid metabolism in cancer cells impact cell structure, signaling, and energy metabolism, making lipid metabolism a potential diagnostic marker and therapeutic target. In this study, we combined PET, desorption electrospray ionization-mass spectrometry (DESI-MS), nonimaging MS, and transcriptomic analyses to interrogate changes in lipid metabolism in a transgenic zebrafish model of oncogenic RAS-driven melanocyte neoplasia progression. Exogenous fatty acid uptake was detected in melanoma tumor nodules by PET using the palmitic acid surrogate tracer 14(R,S)-18F-fluoro-6-thia-heptadecanoic acid ([18F]-FTHA), consistent with upregulation of genes associated with fatty acid uptake found through microarray analysis. DESI-MS imaging revealed that FTHA uptake in tumors was heterogeneous. Transcriptome and lipidome analyses further highlighted dysregulation of glycerophospholipid pathways in melanoma tumor nodules, including increased abundance of phosphatidyl ethanolamine and phosphatidyl choline species, corroborated by DESI-MS, which again revealed heterogeneous phospholipid composition in tumors. Overexpression of the gene encoding lipoprotein lipase (LPL), which was upregulated in zebrafish melanocyte tumor nodules and expressed in the majority of human melanomas, accelerated progression of oncogenic RAS-driven melanocyte neoplasia in zebrafish. Depletion or antagonism of LPL suppressed human melanoma cell growth; this required simultaneous fatty acid synthase (FASN) inhibition when FASN expression was also elevated. Collectively, our findings implicate fatty acid acquisition as a possible therapeutic target in melanoma, and the methods we developed for monitoring fatty acid uptake have potential for diagnosis, patient stratification, and monitoring pharmacologic response. SIGNIFICANCE: These findings demonstrate the translational potential of monitoring fatty acid uptake and identify lipoprotein lipase as a potential therapeutic target in melanoma.


Subject(s)
Fatty Acids/metabolism , Glycerophospholipids/metabolism , Melanocytes/pathology , Melanoma/pathology , Zebrafish/metabolism , Animals , Energy Metabolism , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Humans , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Metabolomics , Microphthalmia-Associated Transcription Factor/genetics , Transcriptome , Tumor Cells, Cultured , Zebrafish/genetics , Zebrafish Proteins/genetics , ras Proteins/genetics , ras Proteins/metabolism
18.
Nat Genet ; 50(9): 1240-1246, 2018 09.
Article in English | MEDLINE | ID: mdl-30127528

ABSTRACT

Childhood high-risk neuroblastomas with MYCN gene amplification are difficult to treat effectively1. This has focused attention on tumor-specific gene dependencies that underlie tumorigenesis and thus provide valuable targets for the development of novel therapeutics. Using unbiased genome-scale CRISPR-Cas9 approaches to detect genes involved in tumor cell growth and survival2-6, we identified 147 candidate gene dependencies selective for MYCN-amplified neuroblastoma cell lines, compared to over 300 other human cancer cell lines. We then used genome-wide chromatin-immunoprecipitation coupled to high-throughput sequencing analysis to demonstrate that a small number of essential transcription factors-MYCN, HAND2, ISL1, PHOX2B, GATA3, and TBX2-are members of the transcriptional core regulatory circuitry (CRC) that maintains cell state in MYCN-amplified neuroblastoma. To disable the CRC, we tested a combination of BRD4 and CDK7 inhibitors, which act synergistically, in vitro and in vivo, with rapid downregulation of CRC transcription factor gene expression. This study defines a set of critical dependency genes in MYCN-amplified neuroblastoma that are essential for cell state and survival in this tumor.


Subject(s)
Gene Expression Regulation, Neoplastic , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Transcription, Genetic , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Disease Models, Animal , Down-Regulation , Female , Gene Amplification , Humans , Mice , Mice, Nude , Transcription Factors/genetics
19.
J Exp Med ; 215(7): 1929-1945, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29941549

ABSTRACT

A substantial subset of patients with T cell acute lymphoblastic leukemia (T-ALL) develops resistance to steroids and succumbs to their disease. JDP2 encodes a bZIP protein that has been implicated as a T-ALL oncogene from insertional mutagenesis studies in mice, but its role in human T-ALL pathogenesis has remained obscure. Here we show that JDP2 is aberrantly expressed in a subset of T-ALL patients and is associated with poor survival. JDP2 is required for T-ALL cell survival, as its depletion by short hairpin RNA knockdown leads to apoptosis. Mechanistically, JDP2 regulates prosurvival signaling through direct transcriptional regulation of MCL1. Furthermore, JDP2 is one of few oncogenes capable of initiating T-ALL in transgenic zebrafish. Notably, thymocytes from rag2:jdp2 transgenic zebrafish express high levels of mcl1 and demonstrate resistance to steroids in vivo. These studies establish JDP2 as a novel oncogene in high-risk T-ALL and implicate overexpression of MCL1 as a mechanism of steroid resistance in JDP2-overexpressing cells.


Subject(s)
Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Repressor Proteins/genetics , Zebrafish Proteins/genetics , Animals , Apoptosis/drug effects , Base Sequence , Cell Proliferation/drug effects , Cell Survival/drug effects , Child, Preschool , Dexamethasone/pharmacology , Disease Models, Animal , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Leukemic/drug effects , Glucocorticoids/pharmacology , Humans , Infant , Mice , Mutagenesis, Insertional/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Transplantation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/metabolism , Response Elements/genetics , Thymocytes/drug effects , Thymocytes/metabolism , Treatment Outcome , Zebrafish , Zebrafish Proteins/metabolism
20.
Cancer Discov ; 8(3): 320-335, 2018 03.
Article in English | MEDLINE | ID: mdl-29284669

ABSTRACT

The amplified MYCN gene serves as an oncogenic driver in approximately 20% of high-risk pediatric neuroblastomas. Here, we show that the family member MYC is a potent transforming gene in a separate subset of high-risk neuroblastoma cases (∼10%), based on (i) its upregulation by focal enhancer amplification or genomic rearrangements leading to enhancer hijacking, and (ii) its ability to transform neuroblastoma precursor cells in a transgenic animal model. The aberrant regulatory elements associated with oncogenic MYC activation include focally amplified distal enhancers and translocation of highly active enhancers from other genes to within topologically associating domains containing the MYC gene locus. The clinical outcome for patients with high levels of MYC expression is virtually identical to that of patients with amplification of the MYCN gene, a known high-risk feature of this disease. Together, these findings establish MYC as a bona fide oncogene in a clinically significant group of high-risk childhood neuroblastomas.Significance: Amplification of the MYCN oncogene is a recognized hallmark of high-risk pediatric neuroblastoma. Here, we demonstrate that MYC is also activated as a potent oncogene in a distinct subset of neuroblastoma cases through either focal amplification of distal enhancers or enhancer hijacking mediated by chromosomal translocation. Cancer Discov; 8(3); 320-35. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 253.


Subject(s)
Enhancer Elements, Genetic , Neuroblastoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Animals, Genetically Modified , Cell Line, Tumor , Child , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , Luminescent Proteins/genetics , N-Myc Proto-Oncogene Protein/genetics , Neoplasms, Experimental/genetics , Neuroblastoma/mortality , Neuroblastoma/pathology , Survival Analysis , Translocation, Genetic , Zebrafish/genetics , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...