Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
bioRxiv ; 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38370799

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor in children and is stratified into three major subgroups. The Sonic hedgehog (SHH) subgroup represents ~30% of all MB cases and has significant survival disparity depending upon TP53 status. Here, we describe the first zebrafish model of SHH MB using CRISPR to mutate ptch1, the primary genetic driver in human SHH MB. These tumors rapidly arise adjacent to the valvula cerebelli and resemble human SHH MB by histology and comparative genomics. In addition, ptch1-deficient MB tumors with loss of tp53 have aggressive tumor histology and significantly worse survival outcomes, comparable to human patients. The simplicity and scalability of the ptch1 MB model makes it highly amenable to CRISPR-based genome editing screens to identify genes required for SHH MB tumor formation in vivo, and here we identify the grk3 kinase as one such target.

3.
iScience ; 26(1): 105737, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36594016

ABSTRACT

Lsd1/Kdm1a functions both as a histone demethylase enzyme and as a scaffold for assembling chromatin modifier and transcription factor complexes to regulate gene expression. The relative contributions of Lsd1's demethylase and scaffolding functions during embryogenesis are not known. Here, we analyze two independent zebrafish lsd1/kdm1a mutant lines and show Lsd1 is required to repress primitive hematopoietic stem cell gene expression. Lsd1 rescue constructs containing point mutations that selectively abrogate its demethylase or scaffolding capacity demonstrate the scaffolding function of Lsd1, not its demethylase activity, is required for repression of gene expression in vivo. Lsd1's SNAG-binding domain mediates its scaffolding function and reinforces a negative feedback loop to repress the expression of SNAG-domain-containing genes during embryogenesis, including gfi1 and snai1/2. Our findings reveal a model in which the SNAG-binding and scaffolding function of Lsd1, and its associated negative feedback loop, provide transient and reversible regulation of gene expression during hematopoietic development.

4.
J Immunol ; 199(5): 1706-1715, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28739882

ABSTRACT

CD79a and CD79b proteins associate with Ig receptors as integral signaling components of the B cell Ag receptor complex. To study B cell development in zebrafish, we isolated orthologs of these genes and performed in situ hybridization, finding that their expression colocalized with IgH-µ in the kidney, which is the site of B cell development. CD79 transgenic lines were made by linking the promoter and upstream regulatory segments of CD79a and CD79b to enhanced GFP to identify B cells, as demonstrated by PCR analysis of IgH-µ expression in sorted cells. We crossed these CD79-GFP lines to a recombination activating gene (Rag)2:mCherry transgenic line to identify B cell development stages in kidney marrow. Initiation of CD79:GFP expression in Rag2:mCherry+ cells and the timing of Ig H and L chain expression revealed simultaneous expression of both IgH-µ- and IgL-κ-chains, without progressing through the stage of IgH-µ-chain alone. Rag2:mCherry+ cells without CD79:GFP showed the highest Rag1 and Rag2 mRNAs compared with CD79a and CD79b:GFP+ B cells, which showed strongly reduced Rag mRNAs. Thus, B cell development in zebrafish does not go through a Raghi CD79+IgH-µ+ pre-B cell stage, different from mammals. After the generation of CD79:GFP+ B cells, decreased CD79 expression occurred upon differentiation to Ig secretion, as detected by alteration from membrane to secreted IgH-µ exon usage, similar to in mammals. This confirmed a conserved role for CD79 in B cell development and differentiation, without the requirement of a pre-B cell stage in zebrafish.


Subject(s)
B-Lymphocytes/physiology , CD79 Antigens/metabolism , Fish Proteins/metabolism , Kidney/physiology , Precursor Cells, B-Lymphoid/physiology , Zebrafish/immunology , Animals , Animals, Genetically Modified , CD79 Antigens/genetics , Cell Differentiation , Cloning, Molecular , DNA-Binding Proteins/genetics , Fish Proteins/genetics , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Lymphocyte Activation , Transgenes/genetics , Zebrafish Proteins/genetics
5.
PLoS One ; 11(12): e0167278, 2016.
Article in English | MEDLINE | ID: mdl-28005909

ABSTRACT

The neural crest (NC) is a stem cell-like embryonic population that is essential for generating and patterning the vertebrate body, including the craniofacial skeleton and peripheral nervous system. Defects in NC development underlie many birth defects and contribute to formation of some of the most malignant cancers in humans, such as melanoma and neuroblastoma. For these reasons, significant research efforts have been expended to identify genes that control NC development, as it is expected to lead to a deeper understanding of the genetic mechanisms controlling vertebrate development and identify new treatments for NC-derived diseases and cancers. However, a number of inconsistencies regarding gene function during NC development have emerged from comparative analyses of gene function between mammalian and non-mammalian systems (chick, frog, zebrafish). This poses a significant barrier to identification of single genes and/or redundant pathways to target in NC diseases. Here, we determine whether technical differences, namely morpholino-based approaches used in non-mammalian systems, could contribute to these discrepancies, by examining the extent to which NC phenotypes in fascin1a (fscn1a) morphant embryos are similar to or different from fscn1a null mutants in zebrafish. Analysis of fscn1a morphants showed that they mimicked early NC phenotypes observed in fscn1a null mutants; however, these embryos also displayed NC migration and derivative phenotypes not observed in null mutants, including accumulation of p53-independent cell death. These data demonstrate that morpholinos can cause seemingly specific NC migration and derivative phenotypes, and thus have likely contributed to the inconsistencies surrounding NC gene function between species. We suggest that comparison of genetic mutants between different species is the most rigorous method for identifying conserved genetic mechanisms controlling NC development and is critical to identify new treatments for NC diseases.


Subject(s)
Microfilament Proteins/metabolism , Morpholinos/metabolism , Neural Crest/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Apoptosis , Body Patterning , Cell Movement , Embryo, Nonmammalian/metabolism , In Situ Hybridization, Fluorescence , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Microscopy, Confocal , Neural Crest/cytology , Phenotype , Pseudopodia/physiology , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
6.
Cell Rep ; 17(5): 1255-1264, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27783941

ABSTRACT

Malignant brain tumors are the leading cause of cancer-related deaths in children. Primitive neuroectodermal tumors of the CNS (CNS-PNETs) are particularly aggressive embryonal tumors of unknown cellular origin. Recent genomic studies have classified CNS-PNETs into molecularly distinct subgroups that promise to improve diagnosis and treatment; however, the lack of cell- or animal-based models for these subgroups prevents testing of rationally designed therapies. Here, we show that a subset of CNS-PNETs co-express oligoneural precursor cell (OPC) markers OLIG2 and SOX10 with coincident activation of the RAS/MAPK (mitogen-activated protein kinase) pathway. Modeling NRAS activation in embryonic OPCs generated malignant brain tumors in zebrafish that closely mimic the human oligoneural/NB-FOXR2 CNS-PNET subgroup by histology and comparative oncogenomics. The zebrafish CNS-PNET model was used to show that MEK inhibitors selectively eliminate Olig2+/Sox10+ CNS-PNET tumors in vivo without impacting normal brain development. Thus, MEK inhibitors represent a promising rationally designed therapy for children afflicted with oligoneural/NB-FOXR2 CNS-PNETs.


Subject(s)
Brain Neoplasms/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasms, Germ Cell and Embryonal/pathology , Neuroectodermal Tumors, Primitive/pathology , Protein Kinase Inhibitors/pharmacology , Stem Cells/metabolism , Animals , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Drug Evaluation, Preclinical , GTP Phosphohydrolases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genome , MAP Kinase Signaling System , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/genetics , Neuroectodermal Tumors, Primitive/drug therapy , Neuroectodermal Tumors, Primitive/genetics , Oncogenes , Protein Kinase Inhibitors/therapeutic use , Stem Cells/drug effects , Zebrafish
7.
Biotechniques ; 60(1): 13-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26757807

ABSTRACT

The zebrafish represents a revolutionary tool in large-scale genetic and small-molecule screens for gene and drug discovery. Transgenic zebrafish are often utilized in these screens. Many transgenic fish lines are maintained in the heterozygous state due to the lethality associated with homozygosity; thus, their progeny must be sorted to ensure a population expressing the transgene of interest for use in screens. Sorting transgenic embryos under a fluorescence microscope is very labor-intensive and demands fine-tuned motor skills. Here we report an efficient transgenic method of utilizing pigmentation rescue of nacre mutant fish for accurate naked-eye identification of both mosaic founders and stable transgenic zebrafish. This was accomplished by co-injecting two constructs with the I-SceI meganuclease enzyme into pigmentless nacre embryos: I-SceI-mitfa:mitfa-I-SceI to rescue the pigmentation and I-SceI-zpromoter:gene-of-interest-I-SceI to express the gene of interest under a zebrafish promoter (zpromoter). Pigmentation rescue reliably predicted transgene integration. Compared with other transgenic techniques, our approach significantly increases the overall percentage of founders and facilitates accurate naked-eye identification of stable transgenic fish, greatly reducing laborious fluorescence microscope sorting and PCR genotyping. Thus, this approach is ideal for generating transgenic fish for large-scale screens.


Subject(s)
Gene Transfer Techniques , Microphthalmia-Associated Transcription Factor/genetics , Pigmentation/genetics , Promoter Regions, Genetic , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Genotype , Green Fluorescent Proteins/genetics , Microscopy, Fluorescence , Zebrafish/genetics , Zebrafish/physiology
8.
Dis Model Mech ; 9(4): 389-400, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26794130

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) is a highly conserved morphogenetic program essential for embryogenesis, regeneration and cancer metastasis. In cancer cells, EMT also triggers cellular reprogramming and chemoresistance, which underlie disease relapse and decreased survival. Hence, identifying compounds that block EMT is essential to prevent or eradicate disseminated tumor cells. Here, we establish a whole-animal-based EMT reporter in zebrafish for rapid drug screening, calledTg(snai1b:GFP), which labels epithelial cells undergoing EMT to producesox10-positive neural crest (NC) cells. Time-lapse and lineage analysis ofTg(snai1b:GFP)embryos reveal that cranial NC cells delaminate from two regions: an early population delaminates adjacent to the neural plate, whereas a later population delaminates from within the dorsal neural tube. TreatingTg(snai1b:GFP)embryos with candidate small-molecule EMT-inhibiting compounds identified TP-0903, a multi-kinase inhibitor that blocked cranial NC cell delamination in both the lateral and medial populations. RNA sequencing (RNA-Seq) analysis and chemical rescue experiments show that TP-0903 acts through stimulating retinoic acid (RA) biosynthesis and RA-dependent transcription. These studies identify TP-0903 as a new therapeutic for activating RAin vivoand raise the possibility that RA-dependent inhibition of EMT contributes to its prior success in eliminating disseminated cancer cells.


Subject(s)
Epithelial Cells/cytology , Epithelial-Mesenchymal Transition/drug effects , Genes, Reporter , Morphogenesis/drug effects , Neural Crest/metabolism , Tretinoin/pharmacology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Shape/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Green Fluorescent Proteins/metabolism , Neural Crest/drug effects , Neural Tube/cytology , Neural Tube/drug effects , Phenotype , Pyrimidines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Transcription, Genetic/drug effects , Zebrafish/embryology , Zebrafish Proteins/metabolism
9.
PLoS Genet ; 11(1): e1004946, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25607881

ABSTRACT

Directional migration of neural crest (NC) cells is essential for patterning the vertebrate embryo, including the craniofacial skeleton. Extensive filopodial protrusions in NC cells are thought to sense chemo-attractive/repulsive signals that provide directionality. To test this hypothesis, we generated null mutations in zebrafish fascin1a (fscn1a), which encodes an actin-bundling protein required for filopodia formation. Homozygous fscn1a zygotic null mutants have normal NC filopodia due to unexpected stability of maternal Fscn1a protein throughout NC development and into juvenile stages. In contrast, maternal/zygotic fscn1a null mutant embryos (fscn1a MZ) have severe loss of NC filopodia. However, only a subset of NC streams display migration defects, associated with selective loss of craniofacial elements and peripheral neurons. We also show that fscn1a-dependent NC migration functions through cxcr4a/cxcl12b chemokine signaling to ensure the fidelity of directional cell migration. These data show that fscn1a-dependent filopodia are required in a subset of NC cells to promote cell migration and NC derivative formation, and that perdurance of long-lived maternal proteins can mask essential zygotic gene functions during NC development.


Subject(s)
Microfilament Proteins/genetics , Neural Crest/growth & development , Pseudopodia/genetics , Zebrafish Proteins/genetics , Zebrafish/growth & development , Animals , Body Patterning/genetics , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Mutation , Neural Crest/cytology , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/biosynthesis
10.
Dev Cell ; 21(3): 492-505, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21920315

ABSTRACT

Endoplasmic reticulum (ER) stress triggers tissue-specific responses that culminate in either cellular adaptation or apoptosis, but the genetic networks distinguishing these responses are not well understood. Here we demonstrate that ER stress induced in the developing zebrafish causes rapid apoptosis in the brain, spinal cord, tail epidermis, lens, and epiphysis. Focusing on the tail epidermis, we uncover an apoptotic response that depends on Puma, but not on p53 or Chop. puma is transcriptionally activated during this ER stress response in a p53-independent manner, and is an essential mediator of epidermal apoptosis. We demonstrate that the p63 transcription factor is upregulated to initiate this apoptotic pathway and directly activates puma transcription in response to ER stress. We also show that a mutation of human Connexin 31, which causes erythrokeratoderma variabilis, induces ER stress and p63-dependent epidermal apoptosis in the zebrafish embryo, thus implicating this pathway in the pathogenesis of inherited disease.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum/metabolism , Epidermis/growth & development , Phosphoproteins/metabolism , Stress, Physiological , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Brefeldin A/pharmacology , Connexins/genetics , Connexins/metabolism , Endoplasmic Reticulum/drug effects , Enzyme Inhibitors/pharmacology , Epidermis/drug effects , Epidermis/metabolism , Erythrokeratodermia Variabilis/genetics , Erythrokeratodermia Variabilis/metabolism , Humans , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Thapsigargin/pharmacology , Transcription, Genetic , Up-Regulation/physiology , Zebrafish Proteins/genetics
11.
Cancer Cell ; 18(4): 353-66, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20951945

ABSTRACT

The molecular events underlying the progression of T-lymphoblastic lymphoma (T-LBL) to acute T-lymphoblastic leukemia (T-ALL) remain elusive. In our zebrafish model, concomitant overexpression of bcl-2 with Myc accelerated T-LBL onset while inhibiting progression to T-ALL. The T-LBL cells failed to invade the vasculature and showed evidence of increased homotypic cell-cell adhesion and autophagy. Further analysis using clinical biopsy specimens revealed autophagy and increased levels of BCL2, S1P1, and ICAM1 in human T-LBL compared with T-ALL. Inhibition of S1P1 signaling in T-LBL cells led to decreased homotypic adhesion in vitro and increased tumor cell intravasation in vivo. Thus, blockade of intravasation and hematologic dissemination in T-LBL is due to elevated S1P1 signaling, increased expression of ICAM1, and augmented homotypic cell-cell adhesion.


Subject(s)
Blood Vessels/pathology , Intercellular Adhesion Molecule-1/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Lysosphingolipid/metabolism , Animals , Animals, Genetically Modified , Autophagy , Blood Vessels/enzymology , Cell Aggregation , Cell Line, Tumor , Cell Movement , Disease Progression , Enzyme Activation , Gene Expression Regulation, Leukemic , Humans , Immunohistochemistry , Intercellular Adhesion Molecule-1/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/genetics , Zebrafish
12.
Cell ; 133(5): 864-77, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18510930

ABSTRACT

Evasion of DNA damage-induced cell death, via mutation of the p53 tumor suppressor or overexpression of prosurvival Bcl-2 family proteins, is a key step toward malignant transformation and therapeutic resistance. We report that depletion or acute inhibition of checkpoint kinase 1 (Chk1) is sufficient to restore gamma-radiation-induced apoptosis in p53 mutant zebrafish embryos. Surprisingly, caspase-3 is not activated prior to DNA fragmentation, in contrast to classical intrinsic or extrinsic apoptosis. Rather, an alternative apoptotic program is engaged that cell autonomously requires atm (ataxia telangiectasia mutated), atr (ATM and Rad3-related) and caspase-2, and is not affected by p53 loss or overexpression of bcl-2/xl. Similarly, Chk1 inhibitor-treated human tumor cells hyperactivate ATM, ATR, and caspase-2 after gamma-radiation and trigger a caspase-2-dependent apoptotic program that bypasses p53 deficiency and excess Bcl-2. The evolutionarily conserved "Chk1-suppressed" pathway defines a novel apoptotic process, whose responsiveness to Chk1 inhibitors and insensitivity to p53 and BCL2 alterations have important implications for cancer therapy.


Subject(s)
Apoptosis , Caspase 2/metabolism , DNA Damage , Protein Kinases/metabolism , Signal Transduction , Zebrafish/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase 3/metabolism , Cell Line, Tumor , Checkpoint Kinase 1 , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/radiation effects , Enzyme Inhibitors/pharmacology , Gamma Rays , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...