1
|
Armistead J, Höpfl S, Goldhausen P, Müller-Hartmann A, Fahle E, Hatzold J, Franzen R, Brodesser S, Radde NE, Hammerschmidt M. A sphingolipid rheostat controls apoptosis versus apical cell extrusion as alternative tumour-suppressive mechanisms. Cell Death Dis 2024; 15:746. [PMID: 39397024 PMCID: PMC11471799 DOI: 10.1038/s41419-024-07134-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024]
Abstract
Evasion of cell death is a hallmark of cancer, and consequently the induction of cell death is a common strategy in cancer treatment. However, the molecular mechanisms regulating different types of cell death are poorly understood. We have formerly shown that in the epidermis of hypomorphic zebrafish hai1a mutant embryos, pre-neoplastic transformations of keratinocytes caused by unrestrained activity of the type II transmembrane serine protease Matriptase-1 heal spontaneously. This healing is driven by Matriptase-dependent increased sphingosine kinase (SphK) activity and sphingosine-1-phosphate (S1P)-mediated keratinocyte loss via apical cell extrusion. In contrast, amorphic hai1afr26 mutants with even higher Matriptase-1 and SphK activity die within a few days. Here we show that this lethality is not due to epidermal carcinogenesis, but to aberrant tp53-independent apoptosis of keratinocytes caused by increased levels of pro-apoptotic C16 ceramides, sphingolipid counterparts to S1P within the sphingolipid rheostat, which severely compromises the epidermal barrier. Mathematical modelling of sphingolipid rheostat homeostasis, combined with in vivo manipulations of components of the rheostat or the ceramide de novo synthesis pathway, indicate that this unexpected overproduction of ceramides is caused by a negative feedback loop sensing ceramide levels and controlling ceramide replenishment via de novo synthesis. Therefore, despite their initial decrease due to increased conversion to S1P, ceramides eventually reach cell death-inducing levels, making transformed pre-neoplastic keratinocytes die even before they are extruded, thereby abrogating the normally barrier-preserving mode of apical live cell extrusion. Our results offer an in vivo perspective of the dynamics of sphingolipid homeostasis and its relevance for epithelial cell survival versus cell death, linking apical cell extrusion and apoptosis. Implications for human carcinomas and their treatments are discussed.
Collapse
Affiliation(s)
- Joy Armistead
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - Sebastian Höpfl
- Institute for Stochastics and Applications, University of Stuttgart, Stuttgart, Germany
| | - Pierre Goldhausen
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany
| | | | - Evelin Fahle
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany
| | - Julia Hatzold
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany
| | - Rainer Franzen
- Max-Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Susanne Brodesser
- Lipidomics/Metabolomics Facility, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Nicole E Radde
- Institute for Stochastics and Applications, University of Stuttgart, Stuttgart, Germany
| | - Matthias Hammerschmidt
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
2
|
Yang CH, Wang YW, Hsu CW, Chung BC. Zebrafish Foxl2l functions in proliferating germ cells for female meiotic entry. Dev Biol 2024; 517:91-99. [PMID: 39341446 DOI: 10.1016/j.ydbio.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Zebrafish sex differentiation is a complicated process and the detailed mechanism has not been fully understood. Here we characterized a transcription factor, Foxl2l, which participates in female oogenesis. We show that it is expressed specifically in proliferating germ cells in juvenile gonads and mature ovaries. We have used CRISPR-Cas9 to generate zebrafish deficient in foxl2l expression. Zebrafish with foxl2l-/- are all males, and this female-to-male sex reversal cannot be reversed by tp53 mutation, indicating this sex reversal is unrelated to cell death. We have generated transgenic fish expressing GFP under the control of foxl2l promoter to track the development of foxl2l + -germ cells; these cells failed to enter meiosis and accumulated as cystic cells in the foxl2l-/- mutant. Our RNA-seq analysis also showed the reduced expression of genes in meiosis and oogenesis among other affected pathways. All together, we show that zebrafish Foxl2l is a nuclear factor controlling the expression of meiotic and oogenic genes, and its deficiency leads to defective meiotic entry and the accumulation of premeiotic germ cells.
Collapse
Affiliation(s)
- Ching-Hsin Yang
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Yan-Wei Wang
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Chen-Wei Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan; National Laboratory Animal Center, National Applied Research Laboratories Taipei, 115, Taiwan
| | - Bon-Chu Chung
- Institute of Molecular Biology, Academia Sinica, Taipei, 115, Taiwan; National Laboratory Animal Center, National Applied Research Laboratories Taipei, 115, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan; Neuroscience and Brain Disease Center, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
3
|
Ferreira PMP, Ramos CLS, Filho JIAB, Conceição MLP, Almeida ML, do Nascimento Rodrigues DC, Porto JCS, de Castro E Sousa JM, Peron AP. Laboratory and physiological aspects of substitute metazoan models for in vivo pharmacotoxicological analysis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03437-5. [PMID: 39298017 DOI: 10.1007/s00210-024-03437-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024]
Abstract
New methods are essential to characterize the performance of substitute procedures for detecting therapeutic action(s) of a chemical or key signal of toxicological events. Herein, it was discussed the applications and advantages of using arthropods, worms, and fishes in pharmacological and/or toxicology assessments. First of all, the illusion of similarity covers many differences between humans and mice, remarkably about liver injury and metabolism of xenobiotics. Using invertebrates, especially earthworms (Eisenia fetida), brine shrimps (Artemia salina, Daphnia magna), and insects (Drosophila melanogaster) and vertebrates as small fishes (Oryzias latipes, Pimephales promelas, Danio rerio) has countless advantages, including fewer ethical conflicts, short life cycle, high reproduction rate, simpler to handle, and less complex anatomy. They can be used to find contaminants in organic matters and water and are easier genetically engineered with orthologous-mutated genes to explore specific proteins involved in proliferative and hormonal disturbances, chemotherapy multidrug resistance, and carcinogenicity. As multicellular embryos, larvae, and mature organisms, they can be tested in bigger-sized replication platforms with 24-, 96-, or 384-multiwell plates as cheaper and faster ways to select hit compounds from drug-like libraries to predict acute, subacute or chronic toxicity, pharmacokinetics, and efficacy parameters of pharmaceutical, cosmetic, and personal care products. Meanwhile, sublethal exposures are designed to identify changes in reproduction, body weight, DNA damages, oxidation, and immune defense responses in earthworms and zebrafishes, and swimming behaviors in A. salina and D. rerio. Behavioral parameters also give specificities on sublethal effects that would not be detected in zebrafishes by OECD protocols.
Collapse
Affiliation(s)
- Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, 64049-550, Brazil.
| | - Carla Lorena Silva Ramos
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, 64049-550, Brazil
| | - José Ivo Araújo Beserra Filho
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, 64049-550, Brazil
| | - Micaely Lorrana Pereira Conceição
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, 64049-550, Brazil
| | - Mateus Lima Almeida
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, 64049-550, Brazil
| | | | - Jhonatas Cley Santos Porto
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, 64049-550, Brazil
| | - João Marcelo de Castro E Sousa
- Toxicological Genetics Research Laboratory (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, 64049-550, Brazil
| | - Ana Paula Peron
- Laboratory of Ecotoxicology (Labecotox), Department of Biodiversity and Nature Conservation, Federal Technological University of Paraná, Campo Mourão, 87301-899, Brazil
| |
Collapse
|
4
|
Shah AN, Leesch F, Lorenzo-Orts L, Grundmann L, Novatchkova M, Haselbach D, Calo E, Pauli A. A dual ribosomal system in the zebrafish soma and germline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610041. [PMID: 39257781 PMCID: PMC11383705 DOI: 10.1101/2024.08.29.610041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Protein synthesis during vertebrate embryogenesis is driven by ribosomes of two distinct origins: maternal ribosomes synthesized during oogenesis and stored in the egg, and somatic ribosomes, produced by the developing embryo after zygotic genome activation (ZGA). In zebrafish, these two ribosome types are expressed from different genomic loci and also differ in their ribosomal RNA (rRNA) sequence. To characterize this dual ribosome system further, we examined the expression patterns of maternal and somatic rRNAs during embryogenesis and in adult tissues. We found that maternal rRNAs are not only expressed during oogenesis but are continuously produced in the zebrafish germline. Proteomic analyses of maternal and somatic ribosomes unveiled differences in core ribosomal protein composition. Most nucleotide differences between maternal and somatic rRNAs are located in the flexible, structurally not resolved expansion segments. Our in vivo data demonstrated that both maternal and somatic ribosomes can be translationally active in the embryo. Using transgenically tagged maternal or somatic ribosome subunits, we experimentally confirm the presence of hybrid 80S ribosomes composed of 40S and 60S subunits from both origins and demonstrate the preferential in vivo association of maternal ribosomes with germline-specific transcripts. Our study identifies a distinct type of ribosomes in the zebrafish germline and thus presents a foundation for future explorations into possible regulatory mechanisms and functional roles of heterogeneous ribosomes.
Collapse
Affiliation(s)
- Arish N. Shah
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Friederike Leesch
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Laura Lorenzo-Orts
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Lorenz Grundmann
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Maria Novatchkova
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - David Haselbach
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Eliezer Calo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States
| | - Andrea Pauli
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| |
Collapse
|
5
|
Casey MJ, Chan PP, Li Q, Zu JF, Jette CA, Kohler M, Myers BR, Stewart RA. A simple and scalable zebrafish model of Sonic hedgehog medulloblastoma. Cell Rep 2024; 43:114559. [PMID: 39078737 PMCID: PMC11404834 DOI: 10.1016/j.celrep.2024.114559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/10/2024] [Accepted: 07/15/2024] [Indexed: 08/07/2024] Open
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 a zebrafish model of SHH MB using CRISPR to create mutant ptch1, the primary genetic driver of human SHH MB. In these animals, tumors rapidly arise in the cerebellum and resemble human SHH MB by histology and comparative onco-genomics. Similar to human patients, MB tumors with loss of both ptch1 and tp53 have aggressive tumor histology and significantly worse survival outcomes. The simplicity and scalability of the ptch1-crispant 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 gene encoding Grk3 kinase as one such target.
Collapse
Affiliation(s)
- Mattie J Casey
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Priya P Chan
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; Primary Children's Hospital, Salt Lake City, UT 84113, USA
| | - Qing Li
- High-Throughput Genomics and Cancer Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Ju-Fen Zu
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Cicely A Jette
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Missia Kohler
- Department of Anatomic Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Benjamin R Myers
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Rodney A Stewart
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
6
|
Beh LK, Shen H. Genotyping Zebrafish Point Mutant by Allele-Specific Blocking PCR. Zebrafish 2024; 21:297-299. [PMID: 38808506 DOI: 10.1089/zeb.2024.0138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
Genotyping zebrafish carrying wild-type, heterozygous, or homozygous copies of a mutant allele is often required for investigating gene specific functions, and is routinely performed to differentiate point mutants. In this study, we describe a modified allele-specific PCR method using an additional blocking primer to promote target sequence amplification while suppressing sequences with single mismatch. Using the tp53m214k point mutant as an example, we show that wild-type, heterozygous, and homozygous zebrafish can be easily distinguished using this simple PCR method, which could be widely adapted for genotyping zebrafish with point mutations or small nucleotide insertions/deletions.
Collapse
Affiliation(s)
- Lih Khiang Beh
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
7
|
Silvius KM, Kendall GC. A Robust Closed-Tube Method for Resolving tp53M214K Genotypes. Zebrafish 2024; 21:250-254. [PMID: 38386541 PMCID: PMC11296207 DOI: 10.1089/zeb.2023.0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
The tp53M214K zebrafish mutant is a versatile platform with which to model a diverse spectrum of human diseases. However, currently available genotyping methods for this mutant require lengthy hands-on processes such as restriction digests and outsourced Sanger sequencing. To address this deficiency, we leveraged high-resolution melting analysis technology in conjunction with a parallel, in-tandem wild-type spike-in approach to develop a robust genotyping protocol capable of discriminating tp53M214K zygosity. In this study, we describe our method in detail. We anticipate that our genotyping protocol will benefit researchers utilizing the tp53M214K zebrafish mutant by offering reliable results with a shorter turnaround time, lower personnel involvement, and higher throughput than traditional methods, thereby decreasing the burden of genotyping and maximizing research efficiency.
Collapse
Affiliation(s)
- Katherine M. Silvius
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Genevieve C. Kendall
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
8
|
Kucinski JP, Calderon D, Kendall GC. Biological and therapeutic insights from animal modeling of fusion-driven pediatric soft tissue sarcomas. Dis Model Mech 2024; 17:dmm050704. [PMID: 38916046 PMCID: PMC11225592 DOI: 10.1242/dmm.050704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024] Open
Abstract
Survival for children with cancer has primarily improved over the past decades due to refinements in surgery, radiation and chemotherapy. Although these general therapies are sometimes curative, the cancer often recurs, resulting in poor outcomes for patients. Fusion-driven pediatric soft tissue sarcomas are genetically defined by chromosomal translocations that create a chimeric oncogene. This distinctive, almost 'monogenic', genetic feature supports the generation of animal models to study the respective diseases in vivo. This Review focuses on a subset of fusion-driven pediatric soft tissue sarcomas that have transgenic animal tumor models, which includes fusion-positive and infantile rhabdomyosarcoma, synovial sarcoma, undifferentiated small round cell sarcoma, alveolar soft part sarcoma and clear cell sarcoma. Studies using the animal models of these sarcomas have highlighted that pediatric cancers require a specific cellular state or developmental stage to drive tumorigenesis, as the fusion oncogenes cause different outcomes depending on their lineage and timing of expression. Therefore, understanding these context-specific activities could identify targetable activities and mechanisms critical for tumorigenesis. Broadly, these cancers show dependencies on chromatin regulators to support oncogenic gene expression and co-opting of developmental pathways. Comparative analyses across lineages and tumor models will further provide biological and therapeutic insights to improve outcomes for these children.
Collapse
Affiliation(s)
- Jack P. Kucinski
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology PhD Program, The Ohio State University, Columbus, OH 43210, USA
| | - Delia Calderon
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology PhD Program, The Ohio State University, Columbus, OH 43210, USA
| | - Genevieve C. Kendall
- Center for Childhood Cancer Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology PhD Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43215, USA
| |
Collapse
|
9
|
Belt AJ, Grant S, Tombes RM, Rothschild SC. Myeloid Targeted Human MLL-ENL and MLL-AF9 Induces cdk9 and bcl2 Expression in Zebrafish Embryos. PLoS Genet 2024; 20:e1011308. [PMID: 38829886 PMCID: PMC11175583 DOI: 10.1371/journal.pgen.1011308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 06/13/2024] [Accepted: 05/19/2024] [Indexed: 06/05/2024] Open
Abstract
Acute myeloid leukemia (AML) accounts for greater than twenty thousand new cases of leukemia annually in the United States. The average five-year survival rate is approximately 30%, pointing to the need for developing novel model systems for drug discovery. In particular, patients with chromosomal rearrangements in the mixed lineage leukemia (MLL) gene have higher relapse rates with poor outcomes. In this study we investigated the expression of human MLL-ENL and MLL-AF9 in the myeloid lineage of zebrafish embryos. We observed an expansion of MLL positive cells and determined these cells colocalized with the myeloid markers spi1b, mpx, and mpeg. In addition, expression of MLL-ENL and MLL-AF9 induced the expression of endogenous bcl2 and cdk9, genes that are often dysregulated in MLL-r-AML. Co-treatment of lyz: MLL-ENL or lyz:MLL-AF9 expressing embryos with the BCL2 inhibitor, Venetoclax, and the CDK9 inhibitor, Flavopiridol, significantly reduced the number of MLL positive cells compared to embryos treated with vehicle or either drug alone. In addition, cotreatment with Venetoclax and Flavopiridol significantly reduced the expression of endogenous mcl1a compared to vehicle, consistent with AML. This new model of MLL-r-AML provides a novel tool to understand the molecular mechanisms underlying disease progression and a platform for drug discovery.
Collapse
MESH Headings
- Zebrafish/genetics
- Zebrafish/embryology
- Animals
- Cyclin-Dependent Kinase 9/genetics
- Cyclin-Dependent Kinase 9/metabolism
- Cyclin-Dependent Kinase 9/antagonists & inhibitors
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Humans
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Sulfonamides/pharmacology
- Piperidines/pharmacology
- Embryo, Nonmammalian
- Flavonoids/pharmacology
- Myeloid Cells/metabolism
- Myeloid Cells/drug effects
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Alex J. Belt
- Life Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Robert M. Tombes
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Sarah C. Rothschild
- Life Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
10
|
Wendt G, Collins JJ. Horizontal gene transfer of a functional cki homolog in the human pathogen Schistosoma mansoni. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.596073. [PMID: 38853947 PMCID: PMC11160599 DOI: 10.1101/2024.05.27.596073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Schistosomes are parasitic flatworms responsible for the neglected tropical disease schistosomiasis, causing devastating morbidity and mortality in the developing world. The parasites are protected by a skin-like tegument, and maintenance of this tegument is controlled by a schistosome ortholog of the tumor suppressor TP53. To understand mechanistically how p53-1 controls tegument production, we identified a cyclin dependent kinase inhibitor homolog (cki) that was co-expressed with p53-1. RNA interference of cki resulted in a hyperproliferation phenotype, that, in combination with p53-1 RNA interference yielded abundant tumor-like growths, indicating that cki and p53-1 are bona fide tumor suppressors in Schistosoma mansoni. Interestingly, cki homologs are widely present throughout parasitic flatworms but evidently absent from their free-living ancestors, suggesting this cki homolog came from an ancient horizontal gene transfer event. This in turn implies that the evolution of parasitism in flatworms may have been aided by a highly unusual means of metazoan genetic inheritance.
Collapse
Affiliation(s)
- George Wendt
- Department of Pharmacology, University of Texas Southwestern Medical Center
| | - James J Collins
- Department of Pharmacology, University of Texas Southwestern Medical Center
| |
Collapse
|
11
|
Prykhozhij SV, Ban K, Brown ZL, Kobar K, Wajnberg G, Fuller C, Chacko S, Lacroix J, Crapoulet N, Midgen C, Shlien A, Malkin D, Berman JN. miR-34a is a tumor suppressor in zebrafish and its expression levels impact metabolism, hematopoiesis and DNA damage. PLoS Genet 2024; 20:e1011290. [PMID: 38805544 PMCID: PMC11166285 DOI: 10.1371/journal.pgen.1011290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 06/11/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024] Open
Abstract
Li-Fraumeni syndrome is caused by inherited TP53 tumor suppressor gene mutations. MicroRNA miR-34a is a p53 target and modifier gene. Interestingly, miR-34 triple-null mice exhibit normal p53 responses and no overt cancer development, but the lack of miR-34 promotes tumorigenesis in cancer-susceptible backgrounds. miR-34 genes are highly conserved and syntenic between zebrafish and humans. Zebrafish miR-34a and miR-34b/c have similar expression timing in development, but miR-34a is more abundant. DNA damage by camptothecin led to p53-dependent induction of miR-34 genes, while miR-34a mutants were adult-viable and had normal DNA damage-induced apoptosis. Nevertheless, miR-34a-/- compound mutants with a gain-of-function tp53R217H/ R217H or tp53-/- mutants were more cancer-prone than tp53 mutants alone, confirming the tumor-suppressive function of miR-34a. Through transcriptomic comparisons at 28 hours post-fertilization (hpf), we characterized DNA damage-induced transcription, and at 8, 28 and 72 hpf we determined potential miR-34a-regulated genes. At 72 hpf, loss of miR-34a enhanced erythrocyte levels and up-regulated myb-positive hematopoietic stem cells. Overexpression of miR-34a suppressed its reporter mRNA, but not p53 target induction, and sensitized injected embryos to camptothecin but not to γ-irradiation.
Collapse
Affiliation(s)
- Sergey V. Prykhozhij
- Children’s Hospital of Eastern Ontario (CHEO) Research Institute and University of Ottawa, Ottawa, Ontario, Canada
| | - Kevin Ban
- Children’s Hospital of Eastern Ontario (CHEO) Research Institute and University of Ottawa, Ottawa, Ontario, Canada
| | - Zane L. Brown
- Dalhousie University Medical School, Halifax, Nova Scotia, Canada
| | - Kim Kobar
- Children’s Hospital of Eastern Ontario (CHEO) Research Institute and University of Ottawa, Ottawa, Ontario, Canada
| | - Gabriel Wajnberg
- Atlantic Cancer Research Institute, Pavillon Hôtel-Dieu, 35 Providence Street, Moncton, NB, Canada
| | - Charlotte Fuller
- HHS McMaster University Medical Centre, Division of Medical Microbiology, Hamilton, Ontario, Canada
| | - Simi Chacko
- Atlantic Cancer Research Institute, Pavillon Hôtel-Dieu, Moncton, New Brunswick, Canada
| | - Jacynthe Lacroix
- Atlantic Cancer Research Institute, Pavillon Hôtel-Dieu, Moncton, New Brunswick, Canada
| | - Nicolas Crapoulet
- Atlantic Cancer Research Institute, Pavillon Hôtel-Dieu, Moncton, New Brunswick, Canada
| | - Craig Midgen
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Adam Shlien
- Genetics and Genome Biology Program, The Hospital for Sick Children, PGCRL, Toronto, Ontario, Canada
| | - David Malkin
- Genetics and Genome Biology Program, The Hospital for Sick Children, PGCRL, Toronto, Ontario, Canada
- Departments of Pediatrics and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jason N. Berman
- Children’s Hospital of Eastern Ontario (CHEO) Research Institute and University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
12
|
Wright SE, Pawlik M, Snyman HN, Haulena M. Review of neoplasia in fish at a large display aquarium, 2005-2021. J Vet Diagn Invest 2024; 36:362-367. [PMID: 38520057 PMCID: PMC11110779 DOI: 10.1177/10406387241241344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2024] Open
Abstract
Fish maintained in managed care may have longer lifespans as a result of advances in veterinary medicine and husbandry and reduced risk of predation. Neoplasia is of increasing interest in managed aquarium populations. However, few studies have systematically evaluated neoplasia in managed fish populations. Our objective in this retrospective study was to review and describe neoplasia diagnosed in fish at a large public display aquarium between 2005 and 2021. Any fish diagnosed with neoplasia on either antemortem or postmortem evaluation during the study period was included, and all medical records, biopsy, and autopsy reports were reviewed. Sixty-two fish met the inclusion criteria; 37 species were included in the study population, most of which were tropical freshwater fish (n = 34 fish). Thirty-two types of neoplasia were identified. Ten fish had benign neoplasms, and 53 fish had malignant neoplasms. The most common neoplasms were of epithelial and neuroectodermal origin. The most common site of tumor origin was the skin. Our data suggest that mesenchymal neoplasms may be more common in cold saltwater fish than in tropical freshwater and saltwater fish. Malignant neoplasms were most commonly diagnosed in the study population and should be a top differential when neoplasms are identified in fish managed under human care. Our study contributes to the overall knowledge of the health of aquarium fish and may aid clinicians in characterizing neoplasia that may be present in fish under human care.
Collapse
Affiliation(s)
- Sarah E. Wright
- Vancouver Aquarium, Vancouver, British Columbia, Canada
- Current address: American Veterinary Medical Association, Schaumburg, IL, USA
| | - Michael Pawlik
- Animal Health Centre–British Columbia Ministry of Agriculture, Abbotsford, British Columbia, Canada
| | - Heindrich N. Snyman
- Animal Health Laboratory–Kemptville, University of Guelph, Kemptville, Ontario, Canada
| | | |
Collapse
|
13
|
Şerifoğlu N, Lopes-Bastos B, Ferreira MG. Lack of telomerase reduces cancer incidence and increases lifespan of zebrafish tp53 M214K mutants. Sci Rep 2024; 14:5382. [PMID: 38443436 PMCID: PMC10914805 DOI: 10.1038/s41598-024-56153-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/01/2024] [Indexed: 03/07/2024] Open
Abstract
Telomerase activity is restricted in humans and telomere attrition occurs in several tissues accompanying natural aging. Critically short telomeres trigger DNA damage responses and activate p53 which leads to apoptosis or replicative senescence. These processes reduce cell proliferation and disrupt tissue homeostasis, thus contributing to systemic aging. Similarly, zebrafish have restricted telomerase expression, and telomeres shorten to critical length during their lifespan. Telomerase-deficient zebrafish (tert -/-) is a premature model of aging that anticipates aging phenotypes due to early telomere shortening. tert -/- zebrafish have impaired cell proliferation, accumulation of DNA damage markers and p53 response. These cellular defects lead to disruption of tissue homeostasis, resulting in premature infertility, gastrointestinal atrophy, sarcopenia and kyphosis. Such consequences contribute to its premature death. Here we reveal a genetic interdependence between tp53 and telomerase function. Mutation of tp53 abrogates premature aging of tert -/- zebrafish, prolonging male fertility and lifespan. However, it does not fully rescue healthspan. tp53mut tert -/- zebrafish retain high levels of inflammation and increased spontaneous cancer incidence. Conversely, loss of telomerase prolongs the lifespan of tp53mut single mutants. Lack of telomerase reduces two-fold the cancer incidence in double mutants and increases lifetime survival. Thus, we observe a reciprocal rescue of tp53mut and tert -/- that ameliorates lifespan but not spontaneous cancer incidence of tp53mut, likely due to higher levels of inflammation.
Collapse
Affiliation(s)
- Naz Şerifoğlu
- Institute for Research on Cancer and Aging of Nice (IRCAN), UMR7284, INSERM U1081, CNRS, Université Cote d'Azur, 06107, Nice, France
| | - Bruno Lopes-Bastos
- Institute for Research on Cancer and Aging of Nice (IRCAN), UMR7284, INSERM U1081, CNRS, Université Cote d'Azur, 06107, Nice, France
| | - Miguel Godinho Ferreira
- Institute for Research on Cancer and Aging of Nice (IRCAN), UMR7284, INSERM U1081, CNRS, Université Cote d'Azur, 06107, Nice, France.
| |
Collapse
|
14
|
Casey MJ, Chan PP, Li Q, Jette CA, Kohler M, Myers BR, Stewart RA. A Simple and Scalable Zebrafish Model of Sonic Hedgehog Medulloblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.03.577834. [PMID: 38370799 PMCID: PMC10871209 DOI: 10.1101/2024.02.03.577834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
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.
Collapse
Affiliation(s)
- Mattie J. Casey
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Priya P. Chan
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
- Primary Children’s Hospital, Salt Lake City, UT 84113, USA
| | - Qing Li
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Cicely A. Jette
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Missia Kohler
- Department of Anatomic Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Benjamin R. Myers
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Rodney A. Stewart
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Lead contact
| |
Collapse
|
15
|
Lawir DF, Soza-Ried C, Iwanami N, Siamishi I, Bylund GO, O Meara C, Sikora K, Kanzler B, Johansson E, Schorpp M, Cauchy P, Boehm T. Antagonistic interactions safeguard mitotic propagation of genetic and epigenetic information in zebrafish. Commun Biol 2024; 7:31. [PMID: 38182651 PMCID: PMC10770094 DOI: 10.1038/s42003-023-05692-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024] Open
Abstract
The stability of cellular phenotypes in developing organisms depends on error-free transmission of epigenetic and genetic information during mitosis. Methylation of cytosine residues in genomic DNA is a key epigenetic mark that modulates gene expression and prevents genome instability. Here, we report on a genetic test of the relationship between DNA replication and methylation in the context of the developing vertebrate organism instead of cell lines. Our analysis is based on the identification of hypomorphic alleles of dnmt1, encoding the DNA maintenance methylase Dnmt1, and pole1, encoding the catalytic subunit of leading-strand DNA polymerase epsilon holoenzyme (Pole). Homozygous dnmt1 mutants exhibit genome-wide DNA hypomethylation, whereas the pole1 mutation is associated with increased DNA methylation levels. In dnmt1/pole1 double-mutant zebrafish larvae, DNA methylation levels are restored to near normal values, associated with partial rescue of mutant-associated transcriptional changes and phenotypes. Hence, a balancing antagonism between DNA replication and maintenance methylation buffers against replicative errors contributing to the robustness of vertebrate development.
Collapse
Affiliation(s)
- Divine-Fondzenyuy Lawir
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Cristian Soza-Ried
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Norimasa Iwanami
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Iliana Siamishi
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Göran O Bylund
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Connor O Meara
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Katarzyna Sikora
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Bioinformatic Unit, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Benoît Kanzler
- Transgenic Mouse Core Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Erik Johansson
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Michael Schorpp
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Pierre Cauchy
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Thomas Boehm
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
16
|
Kent MR, Silvius K, Kucinski J, Calderon D, Kendall GC. Functional Genomics of Novel Rhabdomyosarcoma Fusion-Oncogenes Using Zebrafish. Methods Mol Biol 2024; 2707:23-41. [PMID: 37668903 DOI: 10.1007/978-1-0716-3401-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Clinical sequencing efforts continue to identify novel putative oncogenes with limited strategies to perform functional validation in vivo and study their role in tumorigenesis. Here, we present a pipeline for fusion-driven rhabdomyosarcoma (RMS) in vivo modeling using transgenic zebrafish systems. This strategy originates with novel fusion-oncogenes identified from patient samples that require functional validation in vertebrate systems, integrating these genes into the zebrafish genome, and then characterizing that they indeed drive rhabdomyosarcoma tumor formation. In this scenario, the human form of the fusion-oncogene is inserted into the zebrafish genome to understand if it is an oncogene, and if so, the underlying mechanisms of tumorigenesis. This approach has been successful in our models of infantile rhabdomyosarcoma and alveolar rhabdomyosarcoma, both driven by respective fusion-oncogenes, VGLL2-NCOA2 and PAX3-FOXO1. Our described zebrafish platform is a rapid method to understand the impact of fusion-oncogene activity, divergent and shared fusion-oncogene biology, and whether any analyzed pathways converge for potential clinically actionable targets.
Collapse
Affiliation(s)
- Matthew R Kent
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Katherine Silvius
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Jack Kucinski
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- Molecular, Cellular, and Developmental Biology Ph.D. Program, The Ohio State University, Columbus, OH, USA
| | - Delia Calderon
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- Molecular, Cellular, and Developmental Biology Ph.D. Program, The Ohio State University, Columbus, OH, USA
| | - Genevieve C Kendall
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
17
|
Clevenger T, Paz J, Stafford A, Amos D, Hayes AW. An Evaluation of Zebrafish, an Emerging Model Analyzing the Effects of Toxicants on Cognitive and Neuromuscular Function. Int J Toxicol 2024; 43:46-62. [PMID: 37903286 DOI: 10.1177/10915818231207966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
An emerging alternative to conventional animal models in toxicology research is the zebrafish. Their accelerated development, regenerative capacity, transparent physical appearance, ability to be genetically manipulated, and ease of housing and care make them feasible and efficient experimental models. Nonetheless, their most esteemed asset is their 70% (+) genetic similarity with the human genome, which allows the model to be used in a variety of clinically relevant studies. With these attributes, we propose the zebrafish is an excellent model for analyzing cognitive and neuromuscular responses when exposed to toxicants. Neurocognition can be readily analyzed using visual discrimination, memory and learning, and social behavior testing. Neuromuscular function can be analyzed using techniques such as the startle response, assessment of activity level, and evaluation of critical swimming speed. Furthermore, selectively mutated zebrafish is another novel application of this species in behavioral and pharmacological studies, which can be exploited in toxicological studies. There is a critical need in biomedical research to discover ethical and cost-effective methods to develop new products, including drugs. Through mutagenesis, zebrafish models have become key in meeting this need by advancing the field in numerous areas of biomedical research.
Collapse
Affiliation(s)
| | - Jakob Paz
- Florida College, Temple Terrace, FL, USA
| | | | | | - A Wallace Hayes
- College of Public Health, University of South Florida, Temple Terrace, FL, USA
| |
Collapse
|
18
|
Li Y, Shah RB, Sarti S, Belcher AL, Lee BJ, Gorbatenko A, Nemati F, Yu H, Stanley Z, Rahman M, Shao Z, Silva JM, Zha S, Sidi S. A noncanonical IRAK4-IRAK1 pathway counters DNA damage-induced apoptosis independently of TLR/IL-1R signaling. Sci Signal 2023; 16:eadh3449. [PMID: 38113335 PMCID: PMC11111193 DOI: 10.1126/scisignal.adh3449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023]
Abstract
Interleukin-1 receptor (IL-1R)-associated kinases (IRAKs) are core effectors of Toll-like receptors (TLRs) and IL-1R in innate immunity. Here, we found that IRAK4 and IRAK1 together inhibited DNA damage-induced cell death independently of TLR or IL-1R signaling. In human cancer cells, IRAK4 was activated downstream of ATR kinase in response to double-strand breaks (DSBs) induced by ionizing radiation (IR). Activated IRAK4 then formed a complex with and activated IRAK1. The formation of this complex required the E3 ubiquitin ligase Pellino1, acting structurally but not catalytically, and the activation of IRAK1 occurred independently of extracellular signaling, intracellular TLRs, and the TLR/IL-1R signaling adaptor MyD88. Activated IRAK1 translocated to the nucleus in a Pellino2-dependent manner. In the nucleus, IRAK1 bound to the PIDD1 subunit of the proapoptotic PIDDosome and interfered with platform assembly, thus supporting cell survival. This noncanonical IRAK signaling pathway was also activated in response to other DSB-inducing agents. The loss of IRAK4, of IRAK4 kinase activity, of either Pellino protein, or of the nuclear localization sequence in IRAK1 sensitized p53-mutant zebrafish to radiation. Thus, the findings may lead to strategies for overcoming tumor resistance to conventional cancer treatments.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Richa B. Shah
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Samanta Sarti
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alicia L. Belcher
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian J. Lee
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Andrej Gorbatenko
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Current address: Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Francesca Nemati
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Honglin Yu
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zoe Stanley
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mahbuba Rahman
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhengping Shao
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Jose M. Silva
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shan Zha
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Division of Pediatric Oncology, Hematology and Stem Cell Transplantation, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Samuel Sidi
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
19
|
Oyarbide U, Shah AN, Staton M, Snyderman M, Sapra A, Calo E, Corey SJ. SBDS R126T rescues survival of sbds -/- zebrafish in a dose-dependent manner independently of Tp53. Life Sci Alliance 2023; 6:e202201856. [PMID: 37816584 PMCID: PMC10565674 DOI: 10.26508/lsa.202201856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/12/2023] Open
Abstract
Defects in ribosomal biogenesis profoundly affect organismal development and cellular function, and these ribosomopathies produce a variety of phenotypes. One ribosomopathy, Shwachman-Diamond syndrome (SDS) is characterized by neutropenia, pancreatic exocrine insufficiency, and skeletal anomalies. SDS results from biallelic mutations in SBDS, which encodes a ribosome assembly factor. Some individuals express a missense mutation, SBDS R126T , along with the common K62X mutation. We reported that the sbds-null zebrafish phenocopies much of SDS. We further showed activation of Tp53-dependent pathways before the fish died during the larval stage. Here, we expressed SBDS R126T as a transgene in the sbds -/- background. We showed that one copy of the SBDS R126T transgene permitted the establishment of maternal zygotic sbds-null fish which produced defective embryos with cdkn1a up-regulation, a Tp53 target involved in cell cycle arrest. None survived beyond 3 dpf. However, two copies of the transgene resulted in normal development and lifespan. Surprisingly, neutropenia persisted. The surviving fish displayed suppression of female sex differentiation, a stress response in zebrafish. To evaluate the role of Tp53 in the pathogenesis of sbds -/- fish phenotype, we bred the fish with a DNA binding deficient allele, tp53 M214K Expression of the loss-of-function tp53 M214K did not rescue neutropenia or survival in sbds-null zebrafish. Increased expression of cdkn1a was abrogated in the tp53 M214K/M214K ;sbds -/- fish. We conclude that the amount of SBDSR126T protein is important for development, inactivation of Tp53 fails to rescue neutropenia or survival in the sbds-null background, and cdkn1a up-regulation was dependent on WT tp53 We hypothesize that additional pathways are involved in the pathophysiology of SDS.
Collapse
Affiliation(s)
- Usua Oyarbide
- https://ror.org/03xjacd83 Departments of Cancer Biology and Pediatrics, Cleveland Clinic, Cleveland, OH, USA
| | - Arish N Shah
- Department of Biology and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Morgan Staton
- https://ror.org/03xjacd83 Departments of Cancer Biology and Pediatrics, Cleveland Clinic, Cleveland, OH, USA
| | - Matthew Snyderman
- https://ror.org/03xjacd83 Departments of Cancer Biology and Pediatrics, Cleveland Clinic, Cleveland, OH, USA
| | - Adya Sapra
- https://ror.org/03xjacd83 Departments of Cancer Biology and Pediatrics, Cleveland Clinic, Cleveland, OH, USA
| | - Eliezer Calo
- Department of Biology and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Seth J Corey
- https://ror.org/03xjacd83 Departments of Cancer Biology and Pediatrics, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
20
|
Rothschild SC, Lai G, Tombes RM, Clements WK. Constitutively active CaMKII Drives B lineage acute lymphoblastic leukemia/lymphoma in tp53 mutant zebrafish. PLoS Genet 2023; 19:e1011102. [PMID: 38117861 PMCID: PMC10766190 DOI: 10.1371/journal.pgen.1011102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 01/04/2024] [Accepted: 12/07/2023] [Indexed: 12/22/2023] Open
Abstract
Acute lymphoblastic leukemia/lymphoma (ALL) is the most common pediatric cancer and is a malignancy of T or B lineage lymphoblasts. Dysregulation of intracellular Ca2+ levels has been observed in patients with ALL, leading to improper activation of downstream signaling. Here we describe a new zebrafish model of B ALL, generated by expressing human constitutively active CaMKII (CA-CaMKII) in tp53 mutant lymphocytes. In this model, B cell hyperplasia in the kidney marrow and spleen progresses to overt leukemia/lymphoma, with only 29% of zebrafish surviving the first year of life. Leukemic fish have reduced productive genomic VDJ recombination in addition to reduced expression and improper splicing of ikaros1, a gene often deleted or mutated in patients with B ALL. Inhibiting CaMKII in human pre-B ALL cells induced cell death, further supporting a role for CaMKII in leukemogenesis. This research provides novel insight into the role of Ca2+-directed signaling in lymphoid malignancy and will be useful in understanding disease development and progression.
Collapse
Affiliation(s)
- Sarah C. Rothschild
- Life Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Guanhua Lai
- Pathology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Robert M. Tombes
- Life Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Wilson K. Clements
- Experimental Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
21
|
Bravo P, Liu Y, Draper BW, Marlow FL. Macrophage activation drives ovarian failure and masculinization in zebrafish. SCIENCE ADVANCES 2023; 9:eadg7488. [PMID: 37992158 PMCID: PMC10664988 DOI: 10.1126/sciadv.adg7488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 10/20/2023] [Indexed: 11/24/2023]
Abstract
BMP15 is a conserved regulator of ovarian development and maintenance in vertebrates. In humans, premature ovarian insufficiency is caused by autoimmunity and genetic factors, including mutation of BMP15. The cellular mechanisms underlying ovarian failure caused by BMP15 mutation and immune contributions are not understood. Using zebrafish, we established a causal link between macrophage activation and ovarian failure, which, in zebrafish, causes sex reversal. We define a germline-soma signaling axis that activates macrophages and drives ovarian failure and female-to-male sex reversal. Germline loss of zebrafish Bmp15 impairs oogenesis and initiates this cascade. Single-cell RNA sequencing and genetic analyses implicate ovarian somatic cells that express conserved macrophage-activating ligands as mediators of ovarian failure and sex reversal. Genetic ablation of macrophages or elimination of Csf1Rb ligands, Il34 or Csf1a, delays or blocks premature oocyte loss and sex reversal. The axis identified here provides insight into the cells and pathways governing oocyte and ovary maintenance and potential therapeutic targets to preserve female fertility.
Collapse
Affiliation(s)
- Paloma Bravo
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yulong Liu
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Bruce W. Draper
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Florence L. Marlow
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
22
|
Sun W, Wang M, Zhao J, Zhao S, Zhu W, Wu X, Li F, Liu W, Wang Z, Gao M, Zhang Y, Xu J, Zhang M, Wang Q, Wen Z, Shen J, Zhang W, Huang Z. Sulindac selectively induces autophagic apoptosis of GABAergic neurons and alters motor behaviour in zebrafish. Nat Commun 2023; 14:5351. [PMID: 37660128 PMCID: PMC10475106 DOI: 10.1038/s41467-023-41114-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 08/22/2023] [Indexed: 09/04/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs compose one of the most widely used classes of medications, but the risks for early development remain controversial, especially in the nervous system. Here, we utilized zebrafish larvae to assess the potentially toxic effects of nonsteroidal anti-inflammatory drugs and found that sulindac can selectively induce apoptosis of GABAergic neurons in the brains of zebrafish larvae brains. Zebrafish larvae exhibit hyperactive behaviour after sulindac exposure. We also found that akt1 is selectively expressed in GABAergic neurons and that SC97 (an Akt1 activator) and exogenous akt1 mRNA can reverse the apoptosis caused by sulindac. Further studies showed that sulindac binds to retinoid X receptor alpha (RXRα) and induces autophagy in GABAergic neurons, leading to activation of the mitochondrial apoptotic pathway. Finally, we verified that sulindac can lead to hyperactivity and selectively induce GABAergic neuron apoptosis in mice. These findings suggest that excessive use of sulindac may lead to early neurodevelopmental toxicity and increase the risk of hyperactivity, which could be associated with damage to GABAergic neurons.
Collapse
Affiliation(s)
- Wenwei Sun
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meimei Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jun Zhao
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Shuang Zhao
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wenchao Zhu
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Xiaoting Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Feifei Li
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wei Liu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zhuo Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meng Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Yiyue Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meijia Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Qiang Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, People's Republic of China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518055, China
| | - Juan Shen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Wenqing Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| | - Zhibin Huang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
23
|
Chen J, Baxi K, Lipsitt AE, Hensch NR, Wang L, Sreenivas P, Modi P, Zhao XR, Baudin A, Robledo DG, Bandyopadhyay A, Sugalski A, Challa AK, Kurmashev D, Gilbert AR, Tomlinson GE, Houghton P, Chen Y, Hayes MN, Chen EY, Libich DS, Ignatius MS. Defining function of wild-type and three patient-specific TP53 mutations in a zebrafish model of embryonal rhabdomyosarcoma. eLife 2023; 12:e68221. [PMID: 37266578 PMCID: PMC10322150 DOI: 10.7554/elife.68221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/01/2023] [Indexed: 06/03/2023] Open
Abstract
In embryonal rhabdomyosarcoma (ERMS) and generally in sarcomas, the role of wild-type and loss- or gain-of-function TP53 mutations remains largely undefined. Eliminating mutant or restoring wild-type p53 is challenging; nevertheless, understanding p53 variant effects on tumorigenesis remains central to realizing better treatment outcomes. In ERMS, >70% of patients retain wild-type TP53, yet mutations when present are associated with worse prognosis. Employing a kRASG12D-driven ERMS tumor model and tp53 null (tp53-/-) zebrafish, we define wild-type and patient-specific TP53 mutant effects on tumorigenesis. We demonstrate that tp53 is a major suppressor of tumorigenesis, where tp53 loss expands tumor initiation from <35% to >97% of animals. Characterizing three patient-specific alleles reveals that TP53C176F partially retains wild-type p53 apoptotic activity that can be exploited, whereas TP53P153Δ and TP53Y220C encode two structurally related proteins with gain-of-function effects that predispose to head musculature ERMS. TP53P153Δ unexpectedly also predisposes to hedgehog-expressing medulloblastomas in the kRASG12D-driven ERMS-model.
Collapse
Affiliation(s)
- Jiangfei Chen
- Institute of Environmental Safety and Human Health, Wenzhou Medical UniversityWenzhouChina
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
| | - Kunal Baxi
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Amanda E Lipsitt
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Pediatrics, Division of Hematology Oncology, UT Health Sciences CenterSan AntonioUnited States
| | - Nicole Rae Hensch
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Long Wang
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Prethish Sreenivas
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Paulomi Modi
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Xiang Ru Zhao
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Antoine Baudin
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Biochemistry and Structural Biology, UT Health Sciences CenterSan AntonioUnited States
| | - Daniel G Robledo
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
| | - Abhik Bandyopadhyay
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
| | - Aaron Sugalski
- Department of Pediatrics, Division of Hematology Oncology, UT Health Sciences CenterSan AntonioUnited States
| | - Anil K Challa
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Biology, University of Alabama at BirminghamBirminghamUnited States
| | - Dias Kurmashev
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
| | - Andrea R Gilbert
- Department of Pathology and Laboratory Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Gail E Tomlinson
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Pediatrics, Division of Hematology Oncology, UT Health Sciences CenterSan AntonioUnited States
| | - Peter Houghton
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| | - Yidong Chen
- Department of Population Health Sciences, UT Health Sciences CenterSan AntonioUnited States
| | - Madeline N Hayes
- Developmental and Stem Cell Biology, Hospital for Sick ChildrenTorontoCanada
| | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of WashingtonSeattleUnited States
| | - David S Libich
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Biochemistry and Structural Biology, UT Health Sciences CenterSan AntonioUnited States
| | - Myron S Ignatius
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences CenterSan AntonioUnited States
- Department of Molecular Medicine, UT Health Sciences CenterSan AntonioUnited States
| |
Collapse
|
24
|
Ong AJS, Bladen CE, Tigani TA, Karamalakis AP, Evason KJ, Brown KK, Cox AG. The KEAP1-NRF2 pathway regulates TFEB/TFE3-dependent lysosomal biogenesis. Proc Natl Acad Sci U S A 2023; 120:e2217425120. [PMID: 37216554 PMCID: PMC10235939 DOI: 10.1073/pnas.2217425120] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/17/2023] [Indexed: 05/24/2023] Open
Abstract
The maintenance of redox and metabolic homeostasis is integral to embryonic development. Nuclear factor erythroid 2-related factor 2 (NRF2) is a stress-induced transcription factor that plays a central role in the regulation of redox balance and cellular metabolism. Under homeostatic conditions, NRF2 is repressed by Kelch-like ECH-associated protein 1 (KEAP1). Here, we demonstrate that Keap1 deficiency induces Nrf2 activation and postdevelopmental lethality. Loss of viability is preceded by severe liver abnormalities characterized by an accumulation of lysosomes. Mechanistically, we demonstrate that loss of Keap1 promotes aberrant activation of transcription factor EB (TFEB)/transcription factor binding to IGHM Enhancer 3 (TFE3)-dependent lysosomal biogenesis. Importantly, we find that NRF2-dependent regulation of lysosomal biogenesis is cell autonomous and evolutionarily conserved. These studies identify a role for the KEAP1-NRF2 pathway in the regulation of lysosomal biogenesis and suggest that maintenance of lysosomal homeostasis is required during embryonic development.
Collapse
Affiliation(s)
- Athena Jessica S. Ong
- Peter MacCallum Cancer Centre, Melbourne, VIC3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC3010, Australia
| | - Cerys E. Bladen
- Peter MacCallum Cancer Centre, Melbourne, VIC3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC3010, Australia
| | - Tara A. Tigani
- Peter MacCallum Cancer Centre, Melbourne, VIC3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC3010, Australia
| | - Anthony P. Karamalakis
- Peter MacCallum Cancer Centre, Melbourne, VIC3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC3010, Australia
| | - Kimberley J. Evason
- Division of Anatomic Pathology, Department of Pathology, University of Utah, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT84112, USA
| | - Kristin K. Brown
- Peter MacCallum Cancer Centre, Melbourne, VIC3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC3010, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC3010, Australia
| | - Andrew G. Cox
- Peter MacCallum Cancer Centre, Melbourne, VIC3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC3010, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC3010, Australia
| |
Collapse
|
25
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
26
|
Ajouaou Y, Magnani E, Madakashira B, Jenkins E, Sadler KC. atm Mutation and Oxidative Stress Enhance the Pre-Cancerous Effects of UHRF1 Overexpression in Zebrafish Livers. Cancers (Basel) 2023; 15:cancers15082302. [PMID: 37190230 DOI: 10.3390/cancers15082302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023] Open
Abstract
The ataxia-telangiectasia mutated (atm) gene is activated in response to genotoxic stress and leads to activation of the tp53 tumor suppressor gene which induces either senescence or apoptosis as tumor suppressive mechanisms. Atm also serves non-canonical functions in the response to oxidative stress and chromatin reorganization. We previously reported that overexpression of the epigenetic regulator and oncogene Ubiquitin Like with PHD and Ring Finger Domains 1 (UHRF1) in zebrafish hepatocytes resulted in tp53-dependent hepatocyte senescence, a small liver and larval lethality. We investigated the role of atm on UHRF1-mediated phenotypes by generating zebrafish atm mutants. atm-/- adults were viable but had reduction in fertility. Embryos developed normally but were protected from lethality caused by etoposide or H2O2 exposure and failed to fully upregulate Tp53 targets or oxidative stress response genes in response to these treatments. In contrast to the finding that Tp53 prevents the small liver phenotype caused by UHRF1 overexpression, atm mutation and exposure to H2O2 further reduced the liver size in UHRF1 overexpressing larvae whereas treatment with the antioxidant N-acetyl cysteine suppressed this phenotype. We conclude that UHRF1 overexpression in hepatocytes causes oxidative stress, and that loss of atm further enhances this, triggering elimination of these precancerous cells, leading to a small liver.
Collapse
Affiliation(s)
- Yousra Ajouaou
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Elena Magnani
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Bhavani Madakashira
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Eleanor Jenkins
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| |
Collapse
|
27
|
Song J, Ma J, Liu X, Huang Z, Li L, Li L, Luo L, Ni R, He J. The MRN complex maintains the biliary-derived hepatocytes in liver regeneration through ATR-Chk1 pathway. NPJ Regen Med 2023; 8:20. [PMID: 37024481 PMCID: PMC10079969 DOI: 10.1038/s41536-023-00294-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
When the proliferation of residual hepatocytes is prohibited, biliary epithelial cells (BECs) transdifferentiate into nascent hepatocytes to accomplish liver regeneration. Despite significant interest in transdifferentiation, little is known about the maintenance of nascent hepatocytes in post-injured environments. Here, we perform an N-ethyl-N-nitrosourea (ENU) forward genetic screen and identify a mutant containing a nonsense mutation in the gene nibrin (nbn), which encodes a component of the Mre11-Rad50-Nbn (MRN) complex that activates DNA damage response (DDR). The regenerated hepatocytes cannot be maintained and exhibit apoptosis in the mutant. Mechanistically, the nbn mutation results in the abrogation of ATR-Chk1 signaling and accumulations of DNA damage in nascent hepatocytes, which eventually induces p53-mediated apoptosis. Furthermore, loss of rad50 or mre11a shows similar phenotypes. This study reveals that the activation of DDR by the MRN complex is essential for the survival of BEC-derived hepatocytes, addressing how to maintain nascent hepatocytes in the post-injured environments.
Collapse
Affiliation(s)
- Jingmei Song
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Jianlong Ma
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Xing Liu
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Zhuofu Huang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Lianghui Li
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Linke Li
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Rui Ni
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China.
| | - Jianbo He
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China.
| |
Collapse
|
28
|
Adhish M, Manjubala I. Effectiveness of zebrafish models in understanding human diseases-A review of models. Heliyon 2023; 9:e14557. [PMID: 36950605 PMCID: PMC10025926 DOI: 10.1016/j.heliyon.2023.e14557] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Understanding the detailed mechanism behind every human disease, disorder, defect, and deficiency is a daunting task concerning the clinical diagnostic tools for patients. Hence, a closely resembling living or simulated model is of paramount interest for the development and testing of a probable novel drug for rectifying the conditions pertaining to the various ailments. The animal model that can be easily genetically manipulated to suit the study of the therapeutic motive is an indispensable asset and within the last few decades, the zebrafish models have proven their effectiveness by becoming such potent human disease models with their use being extended to various avenues of research to understand the underlying mechanisms of the diseases. As zebrafish are explored as model animals in understanding the molecular basis and genetics of many diseases owing to the 70% genetic homology between the human and zebrafish genes; new and fascinating facts about the diseases are being surfaced, establishing it as a very powerful tool for upcoming research. These prospective research areas can be explored in the near future using zebrafish as a model. In this review, the effectiveness of the zebrafish as an animal model against several human diseases such as osteoporosis, atrial fibrillation, Noonan syndrome, leukemia, autism spectrum disorders, etc. has been discussed.
Collapse
Affiliation(s)
- Mazumder Adhish
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India
| | - I. Manjubala
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India
| |
Collapse
|
29
|
Watson S, LaVigne CA, Xu L, Surdez D, Cyrta J, Calderon D, Cannon MV, Kent MR, Silvius KM, Kucinski JP, Harrison EN, Murchison W, Rakheja D, Tirode F, Delattre O, Amatruda JF, Kendall GC. VGLL2-NCOA2 leverages developmental programs for pediatric sarcomagenesis. Cell Rep 2023; 42:112013. [PMID: 36656711 PMCID: PMC10054615 DOI: 10.1016/j.celrep.2023.112013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 10/14/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Clinical sequencing efforts are rapidly identifying sarcoma gene fusions that lack functional validation. An example is the fusion of transcriptional coactivators, VGLL2-NCOA2, found in infantile rhabdomyosarcoma. To delineate VGLL2-NCOA2 tumorigenic mechanisms and identify therapeutic vulnerabilities, we implement a cross-species comparative oncology approach with zebrafish, mouse allograft, and patient samples. We find that VGLL2-NCOA2 is sufficient to generate mesenchymal tumors that display features of immature skeletal muscle and recapitulate the human disease. A subset of VGLL2-NCOA2 zebrafish tumors transcriptionally cluster with embryonic somitogenesis and identify VGLL2-NCOA2 developmental programs, including a RAS family GTPase, ARF6. In VGLL2-NCOA2 zebrafish, mouse, and patient tumors, ARF6 is highly expressed. ARF6 knockout suppresses VGLL2-NCOA2 oncogenic activity in cell culture, and, more broadly, ARF6 is overexpressed in adult and pediatric sarcomas. Our data indicate that VGLL2-NCOA2 is an oncogene that leverages developmental programs for tumorigenesis and that reactivation or persistence of ARF6 could represent a therapeutic opportunity.
Collapse
Affiliation(s)
- Sarah Watson
- Institut Curie Research Center, Paris Sciences et Lettres (PSL) Research University, INSERM U830, 75005 Paris, France; Institut Curie, Paris Sciences et Lettres (PSL) Research University, Medical Oncology Department, 75005 Paris, France
| | - Collette A LaVigne
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Didier Surdez
- Institut Curie Research Center, Paris Sciences et Lettres (PSL) Research University, INSERM U830, 75005 Paris, France; Balgrist University Hospital, Faculty of Medicine, University of Zürich (UZH), 8008 Zürich, Switzerland
| | - Joanna Cyrta
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, Department of Pathology, 75005 Paris, France
| | - Delia Calderon
- Center for Childhood Cancer, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA; Molecular, Cellular, and Developmental Biology Ph.D. Program, The Ohio State University, Columbus, OH 43210, USA
| | - Matthew V Cannon
- Center for Childhood Cancer, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Matthew R Kent
- Center for Childhood Cancer, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Katherine M Silvius
- Center for Childhood Cancer, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Jack P Kucinski
- Center for Childhood Cancer, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA; Molecular, Cellular, and Developmental Biology Ph.D. Program, The Ohio State University, Columbus, OH 43210, USA
| | - Emma N Harrison
- Center for Childhood Cancer, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Whitney Murchison
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dinesh Rakheja
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Franck Tirode
- University Lyon, Université Claude Bernard Lyon 1, Cancer Research Center of Lyon, INSERM 1052, CNRS 5286, Centre LéonBérard, 69008 Lyon, France
| | - Olivier Delattre
- Institut Curie Research Center, Paris Sciences et Lettres (PSL) Research University, INSERM U830, 75005 Paris, France; Institut Curie, SIREDO Pediatric Center, 75005 Paris, France; Institut Curie Hospital Group, Unité de Génétique Somatique, 75005 Paris, France
| | - James F Amatruda
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Departments of Pediatrics and Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Genevieve C Kendall
- Center for Childhood Cancer, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43205, USA.
| |
Collapse
|
30
|
Silvius KM, Kendall GC. A robust closed-tube method for resolving tp53 M214K genotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.20.524903. [PMID: 36747876 PMCID: PMC9900746 DOI: 10.1101/2023.01.20.524903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The tp53 M214K zebrafish mutant developed by Berghmans et al 2005 1 is a versatile platform with which to model a diverse spectrum of human diseases. However, currently available genotyping methods for this mutant require lengthy processes such as restriction digests and outsourced Sanger sequencing. To address this deficiency, we leveraged high-resolution melting analysis (HRMA) technology in conjunction with a parallel, in-tandem wildtype spike-in approach to develop a robust genotyping protocol capable of discriminating tp53 M214K zygosity. Here, we describe our method in detail. We anticipate that our genotyping protocol will benefit researchers utilizing the tp53 M214K zebrafish mutant by offering reliable results with a faster turnaround time than conventional approaches.
Collapse
|
31
|
Morgan KJ, Doggett K, Geng F, Mieruszynski S, Whitehead L, Smith KA, Hogan BM, Simons C, Baillie GJ, Molania R, Papenfuss AT, Hall TE, Ober EA, Stainier DYR, Gong Z, Heath JK. ahctf1 and kras mutations combine to amplify oncogenic stress and restrict liver overgrowth in a zebrafish model of hepatocellular carcinoma. eLife 2023; 12:73407. [PMID: 36648336 PMCID: PMC9897728 DOI: 10.7554/elife.73407] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
The nucleoporin (NUP) ELYS, encoded by AHCTF1, is a large multifunctional protein with essential roles in nuclear pore assembly and mitosis. Using both larval and adult zebrafish models of hepatocellular carcinoma (HCC), in which the expression of an inducible mutant kras transgene (krasG12V) drives hepatocyte-specific hyperplasia and liver enlargement, we show that reducing ahctf1 gene dosage by 50% markedly decreases liver volume, while non-hyperplastic tissues are unaffected. We demonstrate that in the context of cancer, ahctf1 heterozygosity impairs nuclear pore formation, mitotic spindle assembly, and chromosome segregation, leading to DNA damage and activation of a Tp53-dependent transcriptional programme that induces cell death and cell cycle arrest. Heterozygous expression of both ahctf1 and ranbp2 (encoding a second nucleoporin), or treatment of heterozygous ahctf1 larvae with the nucleocytoplasmic transport inhibitor, Selinexor, completely blocks krasG12V-driven hepatocyte hyperplasia. Gene expression analysis of patient samples in the liver hepatocellular carcinoma (LIHC) dataset in The Cancer Genome Atlas shows that high expression of one or more of the transcripts encoding the 10 components of the NUP107-160 subcomplex, which includes AHCTF1, is positively correlated with worse overall survival. These results provide a strong and feasible rationale for the development of novel cancer therapeutics that target ELYS function and suggest potential avenues for effective combinatorial treatments.
Collapse
Affiliation(s)
- Kimberly J Morgan
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| | - Karen Doggett
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| | - Fansuo Geng
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| | - Stephen Mieruszynski
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| | - Lachlan Whitehead
- Department of Medical Biology, University of MelbourneParkvilleAustralia
- Centre for Dynamic Imaging, Advanced Technology and Biology Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Kelly A Smith
- Department of Physiology, University of MelbourneParkvilleAustralia
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
| | - Benjamin M Hogan
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
- Peter MacCallum Cancer CentreMelbourneAustralia
| | - Cas Simons
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
- Murdoch Children's Research InstituteParkvilleAustralia
| | - Gregory J Baillie
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
| | - Ramyar Molania
- Department of Medical Biology, University of MelbourneParkvilleAustralia
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Anthony T Papenfuss
- Department of Medical Biology, University of MelbourneParkvilleAustralia
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Thomas E Hall
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
| | - Elke A Ober
- Danish Stem Cell Center, University of CopenhagenCopenhagenDenmark
| | - Didier YR Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Zhiyuan Gong
- Department of Biological Science, National University of SingaporeSingaporeSingapore
| | - Joan K Heath
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| |
Collapse
|
32
|
Bravo P, Liu Y, Draper BW, Marlow FL. Macrophage activation drives ovarian failure and masculinization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.522645. [PMID: 36711702 PMCID: PMC9881905 DOI: 10.1101/2023.01.03.522645] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In humans, premature ovarian insufficiency (POI) is caused by autoimmunity and genetic factors, such as mutation of BMP15, a key ovarian determining gene. The cellular mechanisms associated with ovarian failure caused by BMP15 mutation and immune contributions to the disorder are not understood. BMP15's role in ovarian follicle development is conserved in vertebrates, including zebrafish. Using zebrafish, we established a causal link between macrophage activation and ovarian failure. We identified a germline-somatic gonadal cell-macrophage axis underlying ovarian atresia. Germline loss of Bmp15 triggers this axis that single-cell RNA sequencing and genetic analyses indicate involves activation of ovarian somatic cells that express conserved macrophage-activating ligands. Genetic ablation of macrophages blocks premature oocyte loss. Thus, the axis identified here represents potential therapeutic targets to preserve female fertility.
Collapse
Affiliation(s)
- Paloma Bravo
- Department of Cell, Developmental and Regenerative Biology. Icahn School of Medicine at Mount Sinai; New York, NY, USA
| | - Yulong Liu
- Department of Molecular and Cellular Biology. University of California; Davis, CA, USA
| | - Bruce W. Draper
- Department of Molecular and Cellular Biology. University of California; Davis, CA, USA
| | - Florence L. Marlow
- Department of Cell, Developmental and Regenerative Biology. Icahn School of Medicine at Mount Sinai; New York, NY, USA
| |
Collapse
|
33
|
Zhu W, Shi Y, Zhang C, Peng Y, Wan Y, Xu Y, Liu X, Han B, Zhao S, Kuang Y, Song H, Qiao J. In-frame deletion of SMC5 related with the phenotype of primordial dwarfism, chromosomal instability and insulin resistance. Clin Transl Med 2023; 13:e1007. [PMID: 36627765 PMCID: PMC9832215 DOI: 10.1002/ctm2.1007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 07/16/2022] [Accepted: 07/26/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND SMC5/6 complex plays a vital role in maintaining genome stability, yet the relationship with human diseases has not been described. METHODS SMC5 variation was identified through whole-exome sequencing (WES) and verified by Sanger sequencing. Immunoprecipitation, cytogenetic analysis, fluorescence activated cell sorting (FACS) and electron microscopy were used to elucidate the cellular consequences of patient's cells. smc5 knockout (KO) zebrafish and Smc5K371del knock-in mouse models were generated by CRISPR-Cas9. RNA-seq, quantitative real-time PCR (qPCR), western blot, microquantitative computed tomography (microCT) and histology were used to explore phenotypic characteristics and potential mechanisms of the animal models. The effects of Smc5 knockdown on mitotic clonal expansion (MCE) during adipogenesis were investigated through Oil Red O staining, proliferation and apoptosis assays in vitro. RESULTS We identified a homozygous in-frame deletion of Arg372 in SMC5, one of the core subunits of the SMC5/6 complex, from an adult patient with microcephalic primordial dwarfism, chromosomal instability and insulin resistance. SMC5 mutation disrupted its interaction with its interacting protein NSMCE2, leading to defects in DNA repair and chromosomal instability in patient fibroblasts. Smc5 KO zebrafish showed microcephaly, short length and disturbed glucose metabolism. Smc5 depletion triggers a p53-related apoptosis, as concomitant deletion of the p53 rescued growth defects phenotype in zebrafish. An smc5K371del knock-in mouse model exhibited high mortality, severe growth restriction and fat loss. In 3T3-L1 cells, the knockdown of smc5 results in impaired MCE, a crucial step in adipogenesis. This finding implies that defective cell survival and differentiation is an important mechanism linking growth disorders and metabolic homeostasis imbalance.
Collapse
Affiliation(s)
- Wenjiao Zhu
- Department of EndocrinologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuanping Shi
- Department of EndocrinologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Changrun Zhang
- Department of EndocrinologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yajie Peng
- Department of EndocrinologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yueyue Wan
- Department of Molecular Diagnostics & EndocrinologyThe Core Laboratory in Medical Center of Clinical ResearchShanghai Ninth People's HospitalState Key Laboratory of Medical GenomicsShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yue Xu
- Department of EndocrinologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xuemeng Liu
- Department of EndocrinologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bing Han
- Department of EndocrinologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuangxia Zhao
- Department of Molecular Diagnostics & EndocrinologyThe Core Laboratory in Medical Center of Clinical ResearchShanghai Ninth People's HospitalState Key Laboratory of Medical GenomicsShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yanping Kuang
- Department of Assisted ReproductionShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Huaidong Song
- Department of Molecular Diagnostics & EndocrinologyThe Core Laboratory in Medical Center of Clinical ResearchShanghai Ninth People's HospitalState Key Laboratory of Medical GenomicsShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Qiao
- Department of EndocrinologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
34
|
Yi ZN, Chen XK, Ma ACH. Modeling leukemia with zebrafish (Danio rerio): Towards precision medicine. Exp Cell Res 2022; 421:113401. [PMID: 36306826 DOI: 10.1016/j.yexcr.2022.113401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 12/29/2022]
Abstract
Leukemia is a type of blood cancer characterized by high genetic heterogeneity and fatality. While chemotherapy remains the primary form of treatment for leukemia, its effectiveness was profoundly diminished by the genetic heterogeneity and cytogenetic abnormalities of leukemic cells. Therefore, there is an unmet need to develop precision medicine for leukemia with distinct genetic backgrounds. Zebrafish (Danio rerio), a freshwater fish with exceptional feasibility in genome editing, is a powerful tool for rapid human cancer modeling. In the past decades, zebrafish have been adopted in modeling human leukemia, exploring the molecular mechanisms of underlying genetic abnormalities, and discovering novel therapeutic agents. Although many recurrent mutations of leukemia have been modeled in zebrafish for pathological study and drug discovery, its great potential in leukemia modeling was not yet fully exploited, particularly in precision medicine. In this review, we evaluated the current zebrafish models of leukemia/pre-leukemia and genetic techniques and discussed the potential of zebrafish models with novel techniques, which may contribute to the development of zebrafish as a disease model for precision medicine in treating leukemia.
Collapse
Affiliation(s)
- Zhen-Ni Yi
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xiang-Ke Chen
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Alvin Chun-Hang Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
35
|
Potts KS, Cameron RC, Metidji A, Ghazale N, Wallace L, Leal-Cervantes AI, Palumbo R, Barajas JM, Gupta V, Aluri S, Pradhan K, Myers JA, McKinstry M, Bai X, Choudhary GS, Shastri A, Verma A, Obeng EA, Bowman TV. Splicing factor deficits render hematopoietic stem and progenitor cells sensitive to STAT3 inhibition. Cell Rep 2022; 41:111825. [PMID: 36516770 PMCID: PMC9994853 DOI: 10.1016/j.celrep.2022.111825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 10/01/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) sustain lifelong hematopoiesis. Mutations of pre-mRNA splicing machinery, especially splicing factor 3b, subunit 1 (SF3B1), are early lesions found in malignancies arising from HSPC dysfunction. However, why splicing factor deficits contribute to HSPC defects remains incompletely understood. Using zebrafish, we show that HSPC formation in sf3b1 homozygous mutants is dependent on STAT3 activation. Clinically, mutations in SF3B1 are heterozygous; thus, we explored if targeting STAT3 could be a vulnerability in these cells. We show that SF3B1 heterozygosity confers heightened sensitivity to STAT3 inhibition in zebrafish, mouse, and human HSPCs. Cells carrying mutations in other splicing factors or treated with splicing modulators are also more sensitive to STAT3 inhibition. Mechanistically, we illustrate that STAT3 inhibition exacerbates aberrant splicing in SF3B1 mutant cells. Our findings reveal a conserved vulnerability of splicing factor mutant HSPCs that could allow for their selective targeting in hematologic malignancies.
Collapse
Affiliation(s)
- Kathryn S Potts
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rosannah C Cameron
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Amina Metidji
- Department of Oncology, St. Jude's Children Research Hospital, Memphis, TN 38105, USA
| | - Noura Ghazale
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - LaShanale Wallace
- Department of Oncology, St. Jude's Children Research Hospital, Memphis, TN 38105, USA
| | - Ana I Leal-Cervantes
- Department of Oncology, St. Jude's Children Research Hospital, Memphis, TN 38105, USA
| | - Reid Palumbo
- Department of Oncology, St. Jude's Children Research Hospital, Memphis, TN 38105, USA
| | - Juan Martin Barajas
- Department of Oncology, St. Jude's Children Research Hospital, Memphis, TN 38105, USA
| | - Varun Gupta
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Srinivas Aluri
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - Kith Pradhan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - Jacquelyn A Myers
- Department of Oncology, St. Jude's Children Research Hospital, Memphis, TN 38105, USA
| | - Mia McKinstry
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Xiaoying Bai
- Department of Obstetrics and Gynecology, University of Texas, Dallas, TX, USA
| | - Gaurav S Choudhary
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - Aditi Shastri
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; Montefiore Einstein Cancer Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - Amit Verma
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; Montefiore Einstein Cancer Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - Esther A Obeng
- Department of Oncology, St. Jude's Children Research Hospital, Memphis, TN 38105, USA.
| | - Teresa V Bowman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; Montefiore Einstein Cancer Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA.
| |
Collapse
|
36
|
van de Beek I, Glykofridis IE, Wagner A, den Toom DT, Bongers EMHF, van Leenders GJLH, Johannesma PC, Meijers‐Heijboer HEJ, Wolthuis RMF, van Steensel MAM, Dubbink HJ, Houweling AC. Combined germline pathogenic variants in FLCN and TP53 are associated with early onset renal cell carcinoma and brain tumors. Mol Genet Genomic Med 2022; 11:e2098. [PMID: 36382415 PMCID: PMC9938753 DOI: 10.1002/mgg3.2098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND We present a family consisting of a father and his two children with an exceptional phenotype of childhood renal cell carcinoma and brain tumors. Extensive genetic testing revealed two inherited tumor predisposition syndromes in all three family members: Birt-Hogg-Dubé syndrome and Li-Fraumeni syndrome. The corresponding genes (FLCN and TP53) are both located on the short arm of chromosome 17. METHODS We describe the phenotype and performed single nucleotide polymorphism (SNP)-based loss of heterozygosity (LOH) analysis of the tumors. RESULTS All examined tumors showed somatic loss of the wild-type alleles of both FLCN and TP53. CONCLUSIONS We hypothesize that a synergistic effect of both mutations caused the unusual phenotype of childhood renal cell carcinoma in this family. This family emphasizes the importance of further genetic testing if a tumor develops at an unexpected young age in an inherited cancer predisposition syndrome.
Collapse
Affiliation(s)
- Irma van de Beek
- Department of Human GeneticsAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Iris E. Glykofridis
- Department of Human Genetics, Cancer Center AmsterdamAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Anja Wagner
- Department of Clinical GeneticsUniversity Medical Center Rotterdam, Erasmus MC Cancer InstituteRotterdamthe Netherlands
| | - Dorine T. den Toom
- Department of PathologyUniversity Medical Center Rotterdam, Erasmus MC Cancer InstituteRotterdamthe Netherlands
| | | | - Geert J. L. H. van Leenders
- Department of PathologyUniversity Medical Center Rotterdam, Erasmus MC Cancer InstituteRotterdamthe Netherlands
| | - Paul C. Johannesma
- Department of PulmonologyAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | | | - Rob M. F. Wolthuis
- Department of Human Genetics, Cancer Center AmsterdamAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Maurice A. M. van Steensel
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore,Singapore Skin Research Institute, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Hendrikus J. Dubbink
- Department of PathologyUniversity Medical Center Rotterdam, Erasmus MC Cancer InstituteRotterdamthe Netherlands
| | - Arjan C. Houweling
- Department of Human GeneticsAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| |
Collapse
|
37
|
Wild-type and cancer-prone zebrafish exhibit distinct gut microbial diversity and differential anti-inflammatory response upon infection. J Biosci 2022. [DOI: 10.1007/s12038-022-00302-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
38
|
Costa JCD, Souza SSD, Val AL. Impact of high temperature, CO 2 and parasitic infection on inflammation, immunodepression and programmed cell death in Colossoma macropomum at the transcriptional level. Microb Pathog 2022; 172:105804. [PMID: 36179975 DOI: 10.1016/j.micpath.2022.105804] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 10/31/2022]
Abstract
The production of tambaqui Colossoma macropomum has recently reached a milestone, being considered the main native species produced in South American continental waters. Despite the importance of this fish, its immunity is poorly understood, and global warming could pose severe risks to its health as increasing water temperature leads to an increase in the incidence of parasitic diseases. In an experimental context based on the high-emission scenario of the 5th Intergovernmental Panel on Climate Change (IPCC) report, we evaluated the synergistic effect of exposure to the extreme climate change scenario (RCP8.5) during two exposure periods (7 and 30 days) and two levels of parasitism by monogeneans (low and high). The goal was to understand how the tambaqui immune system will react to this challenge. To achieve this goal, we analyzed the expression of nine immunity-related genes (jak3, stat3, il-10, socs1, casp1, il-1β, tp53, bcl2, and hif-1α) in the spleen. Our main findings showed downregulation in the jak3/stat3 pathway, genes related to the control of inflammation and apoptosis, in addition to upregulation of proinflammatory genes and those related to pyroptosis during the first 7 days of exposure to the extreme climate scenario, also indicating a stage of immunodepression in these animals. After 30 days of exposure, all genes tended to return to similar levels in the current scenario, possibly due to the decrease in parasite load caused by chronic exposure to the extreme scenario. Our data strongly suggest that the increase in parasitism intensity caused by the extreme climate change scenario is responsible for disturbances in the host's immune system. However, more studies are needed to clarify this poorly understood cascade of events.
Collapse
Affiliation(s)
- Jaqueline Custódio da Costa
- Graduate Program in Genetics, Conservation and Evolutionary Biology (PPG-GCBEv), Laboratory of Ecophysiology and Molecular Evolution (LEEM), Brazilian National Institute for Research of the Amazon (INPA), 69067-375, Manaus, Amazonas, Brazil.
| | - Samara Silva de Souza
- Graduate Program in Genetics, Conservation and Evolutionary Biology (PPG-GCBEv), Laboratory of Ecophysiology and Molecular Evolution (LEEM), Brazilian National Institute for Research of the Amazon (INPA), 69067-375, Manaus, Amazonas, Brazil
| | - Adalberto Luis Val
- Graduate Program in Genetics, Conservation and Evolutionary Biology (PPG-GCBEv), Laboratory of Ecophysiology and Molecular Evolution (LEEM), Brazilian National Institute for Research of the Amazon (INPA), 69067-375, Manaus, Amazonas, Brazil
| |
Collapse
|
39
|
Dudziak K, Nowak M, Sozoniuk M. One Host-Multiple Applications: Zebrafish ( Danio rerio) as Promising Model for Studying Human Cancers and Pathogenic Diseases. Int J Mol Sci 2022; 23:10255. [PMID: 36142160 PMCID: PMC9499349 DOI: 10.3390/ijms231810255] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, zebrafish (ZF) has been increasingly applied as a model in human disease studies, with a particular focus on cancer. A number of advantages make it an attractive alternative for mice widely used so far. Due to the many advantages of zebrafish, modifications can be based on different mechanisms and the induction of human disease can take different forms depending on the research goal. Genetic manipulation, tumor transplantation, or injection of the pathogen are only a few examples of using ZF as a model. Most of the studies are conducted in order to understand the disease mechanism, monitor disease progression, test new or alternative therapies, and select the best treatment. The transplantation of cancer cells derived from patients enables the development of personalized medicine. To better mimic a patient's body environment, immune-deficient models (SCID) have been developed. A lower immune response is mostly generated by genetic manipulation but also by irradiation or dexamethasone treatment. For many studies, using SCID provides a better chance to avoid cancer cell rejection. In this review, we describe the main directions of using ZF in research, explain why and how zebrafish can be used as a model, what kind of limitations will be met and how to overcome them. We collected recent achievements in this field, indicating promising perspectives for the future.
Collapse
Affiliation(s)
- Karolina Dudziak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Michał Nowak
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| | - Magdalena Sozoniuk
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| |
Collapse
|
40
|
Ercc2/Xpd deficiency results in failure of digestive organ growth in zebrafish with elevated nucleolar stress. iScience 2022; 25:104957. [PMID: 36065184 PMCID: PMC9440294 DOI: 10.1016/j.isci.2022.104957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/23/2022] [Accepted: 08/12/2022] [Indexed: 12/09/2022] Open
Abstract
Mutations in ERCC2/XPD helicase, an important component of the TFIIH complex, cause distinct human genetic disorders which exhibit various pathological features. However, the molecular mechanisms underlying many symptoms remain elusive. Here, we have shown that Ercc2/Xpd deficiency in zebrafish resulted in hypoplastic digestive organs with normal bud initiation but later failed to grow. The proliferation of intestinal endothelial cells was impaired in ercc2/xpd mutants, and mitochondrial abnormalities, autophagy, and inflammation were highly induced. Further studies revealed that these abnormalities were associated with the perturbation of rRNA synthesis and nucleolar stress in a p53-independent manner. As TFIIH has only been implicated in RNA polymerase I-dependent transcription in vitro, our results provide the first evidence for the connection between Ercc2/Xpd and rRNA synthesis in an animal model that recapitulates certain key characteristics of ERCC2/XPD-related human genetic disorders, and will greatly advance our understanding of the molecular pathogenesis of these diseases. Ercc2/Xpd deficiency results in failure of digestive organ growth in zebrafish Ercc2/Xpd-deficient intestinal endothelial cells exhibit impaired proliferation Mitochondrial abnormalities, autophagy, and inflammation are highly induced rRNA synthesis perturbation leads to nucleolar stress in a p53-independent manner
Collapse
|
41
|
Basheer F, Dhar P, Samarasinghe RM. Zebrafish Models of Paediatric Brain Tumours. Int J Mol Sci 2022; 23:9920. [PMID: 36077320 PMCID: PMC9456103 DOI: 10.3390/ijms23179920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Paediatric brain cancer is the second most common childhood cancer and is the leading cause of cancer-related deaths in children. Despite significant advancements in the treatment modalities and improvements in the 5-year survival rate, it leaves long-term therapy-associated side effects in paediatric patients. Addressing these impairments demands further understanding of the molecularity and heterogeneity of these brain tumours, which can be demonstrated using different animal models of paediatric brain cancer. Here we review the use of zebrafish as potential in vivo models for paediatric brain tumour modelling, as well as catalogue the currently available zebrafish models used to study paediatric brain cancer pathophysiology, and discuss key findings, the unique attributes that these models add, current challenges and therapeutic significance.
Collapse
Affiliation(s)
- Faiza Basheer
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| | - Poshmaal Dhar
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| | - Rasika M. Samarasinghe
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| |
Collapse
|
42
|
Wang J, Thomas HR, Chen Y, Percival SM, Waldrep SC, Ramaker RC, Thompson RG, Cooper SJ, Chong Z, Parant JM. Reduced sister chromatid cohesion acts as a tumor penetrance modifier. PLoS Genet 2022; 18:e1010341. [PMID: 35994499 PMCID: PMC9436123 DOI: 10.1371/journal.pgen.1010341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/01/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sister chromatid cohesion (SCC) is an important process in chromosome segregation. ESCO2 is essential for establishment of SCC and is often deleted/altered in human cancers. We demonstrate that esco2 haploinsufficiency results in reduced SCC and accelerates the timing of tumor onset in both zebrafish and mouse p53 heterozygous null models, but not in p53 homozygous mutant or wild-type animals. These data indicate that esco2 haploinsufficiency accelerates tumor onset in a loss of heterozygosity (LOH) sensitive background. Analysis of The Cancer Genome Atlas (TCGA) confirmed ESCO2 deficient tumors have elevated number of LOH events throughout the genome. Further, we demonstrated heterozygous loss of sgo1, important in maintaining SCC, also results in reduced SCC and accelerated tumor formation in a p53 heterozygous background. Surprisingly, while we did observe elevated levels of chromosome missegregation and micronuclei formation in esco2 heterozygous mutant animals, this chromosomal instability did not contribute to the accelerated tumor onset in a p53 heterozygous background. Interestingly, SCC also plays a role in homologous recombination, and we did observe elevated levels of mitotic recombination derived p53 LOH in tumors from esco2 haploinsufficient animals; as well as elevated levels of mitotic recombination throughout the genome of human ESCO2 deficient tumors. Together these data suggest that reduced SCC contributes to accelerated tumor penetrance through elevated mitotic recombination. Tumorigenesis often involves the inactivation of tumor suppressor genes. This often encompasses an inactivation mutation in one allele and loss of the other wild-type allele, referred to as loss of heterozygosity (LOH). The rate at which the cells lose the wild-type allele can influence the timing of tumor onset, and therefore an indicator of a patient’s risk of cancer. Factors that influence this process could be used as a predictive indicator of cancer risk, however these factors are still unclear. We demonstrate that partial impairment of sister chromatid cohesion (SCC), a fundamental component of the chromosome segregation in mitosis and homologous recombination repair, enhanced tumorigenesis. Our data suggest this is through elevated levels of mitotic recombination derived p53 LOH. This study emphasizes the importance of understanding how impaired SCC, mitotic recombination rates, and LOH rates influence cancer risk.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Holly R. Thomas
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Yu Chen
- Department of Genetics, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
- Informatics Institute, University of Alabama at Birmingham Heersink School of Medicine, Alabama, United States of America
| | - Stefanie M. Percival
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Stephanie C. Waldrep
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Ryne C. Ramaker
- Hudson Alpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Robert G. Thompson
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Sara J. Cooper
- Hudson Alpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Zechen Chong
- Department of Genetics, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
- Informatics Institute, University of Alabama at Birmingham Heersink School of Medicine, Alabama, United States of America
| | - John M. Parant
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
43
|
Ran R, Li L, Shi Z, Liu G, Jiang H, Fang L, Xu T, Huang J, Chen W, Chen Y. Disruption of
tp53
leads to cutaneous nevus and melanoma formation in
Xenopus tropicalis. Mol Oncol 2022; 16:3554-3567. [PMID: 35981147 PMCID: PMC9533689 DOI: 10.1002/1878-0261.13301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/22/2022] [Accepted: 08/05/2022] [Indexed: 11/10/2022] Open
Abstract
In humans, germline TP53 mutations predispose carriers to a wide spectrum of cancers, which is known as Li–Fraumeni syndrome (LFS). To date, the association of melanomas with LFS remains unestablished. No melanomas have been reported in any P53‐modified mouse models either. In this study, we show that targeted disruption of P53 at the DNA‐binding domain in Xenopus tropicalis recapitulates LFS, with the formation of soft‐tissue sarcomas and pancreatic ductal adenocarcinoma. Interestingly, 19% of the 14‐month‐old tp53Δ7/Δ7 homozygotes and 18% of tp53+/Δ7 heterozygotes spontaneously developed small nevi and non‐invasive melanomas. Large invasive melanomas were also observed in other older homozygous mutants, with about 7.9% penetrance. Our data suggest that more dermatologic investigation of LFS patients should be able to settle the association of melanoma with LFS in epidemiology. Our model is also valuable for further investigation of the molecular mechanism underlying melanoma progression upon germline alteration of the tp53 locus.
Collapse
Affiliation(s)
- Rensen Ran
- School of Life Science and Technology Harbin Institute of Technology Harbin China
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| | - Lanxin Li
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| | - Zhaoying Shi
- School of Life Science and Technology Harbin Institute of Technology Harbin China
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| | - Guanghui Liu
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| | - Hao Jiang
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| | - Liangchen Fang
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| | - Tingting Xu
- School of Medical Technology and Engineering Fujian Medical University Fuzhou China
| | - Jixuan Huang
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| | - Weiqi Chen
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| | - Yonglong Chen
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology Shenzhen China
| |
Collapse
|
44
|
Darp R, Vittoria MA, Ganem NJ, Ceol CJ. Oncogenic BRAF induces whole-genome doubling through suppression of cytokinesis. Nat Commun 2022; 13:4109. [PMID: 35840569 PMCID: PMC9287415 DOI: 10.1038/s41467-022-31899-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
Melanomas and other solid tumors commonly have increased ploidy, with near-tetraploid karyotypes being most frequently observed. Such karyotypes have been shown to arise through whole-genome doubling events that occur during early stages of tumor progression. The generation of tetraploid cells via whole-genome doubling is proposed to allow nascent tumor cells the ability to sample various pro-tumorigenic genomic configurations while avoiding the negative consequences that chromosomal gains or losses have in diploid cells. Whereas a high prevalence of whole-genome doubling events has been established, the means by which whole-genome doubling arises is unclear. Here, we find that BRAFV600E, the most common mutation in melanomas, can induce whole-genome doubling via cytokinesis failure in vitro and in a zebrafish melanoma model. Mechanistically, BRAFV600E causes decreased activation and localization of RhoA, a critical cytokinesis regulator. BRAFV600E activity during G1/S phases of the cell cycle is required to suppress cytokinesis. During G1/S, BRAFV600E activity causes inappropriate centriole amplification, which is linked in part to inhibition of RhoA and suppression of cytokinesis. Together these data suggest that common abnormalities of melanomas linked to tumorigenesis - amplified centrosomes and whole-genome doubling events - can be induced by oncogenic BRAF and other mutations that increase RAS/MAPK pathway activity.
Collapse
Affiliation(s)
- Revati Darp
- University of Massachusetts Chan Medical School, Program in Molecular Medicine, Worcester, MA, USA
- University of Massachusetts Chan Medical School, Department of Molecular, Cellular and Cancer Biology, Worcester, MA, USA
| | - Marc A Vittoria
- Departments of Pharmacology and Experimental Therapeutics and Medicine, Division of Hematology and Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Neil J Ganem
- Departments of Pharmacology and Experimental Therapeutics and Medicine, Division of Hematology and Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Craig J Ceol
- University of Massachusetts Chan Medical School, Program in Molecular Medicine, Worcester, MA, USA.
- University of Massachusetts Chan Medical School, Department of Molecular, Cellular and Cancer Biology, Worcester, MA, USA.
| |
Collapse
|
45
|
Russo I, Sartor E, Fagotto L, Colombo A, Tiso N, Alaibac M. The Zebrafish model in dermatology: an update for clinicians. Discov Oncol 2022; 13:48. [PMID: 35713744 PMCID: PMC9206045 DOI: 10.1007/s12672-022-00511-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/08/2022] [Indexed: 11/04/2022] Open
Abstract
Recently, the zebrafish has been established as one of the most important model organisms for medical research. Several studies have proved that there is a high level of similarity between human and zebrafish genomes, which encourages the use of zebrafish as a model for understanding human genetic disorders, including cancer. Interestingly, zebrafish skin shows several similarities to human skin, suggesting that this model organism is particularly suitable for the study of neoplastic and inflammatory skin disorders. This paper appraises the specific characteristics of zebrafish skin and describes the major applications of the zebrafish model in dermatological research.
Collapse
Affiliation(s)
- Irene Russo
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy
| | - Emma Sartor
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy
| | - Laura Fagotto
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy
| | - Anna Colombo
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy
| | - Natascia Tiso
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35131, Padua, Italy
| | - Mauro Alaibac
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy.
| |
Collapse
|
46
|
Disease modeling by efficient genome editing using a near PAM-less base editor in vivo. Nat Commun 2022; 13:3435. [PMID: 35701478 PMCID: PMC9198099 DOI: 10.1038/s41467-022-31172-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 06/08/2022] [Indexed: 12/18/2022] Open
Abstract
Base Editors are emerging as an innovative technology to introduce point mutations in complex genomes. So far, the requirement of an NGG Protospacer Adjacent Motif (PAM) at a suitable position often limits the base editing possibility to model human pathological mutations in animals. Here we show that, using the CBE4max-SpRY variant recognizing nearly all PAM sequences, we could introduce point mutations for the first time in an animal model with high efficiency, thus drastically increasing the base editing possibilities. With this near PAM-less base editor we could simultaneously mutate several genes and we developed a co-selection method to identify the most edited embryos based on a simple visual screening. Finally, we apply our method to create a zebrafish model for melanoma predisposition based on the simultaneous base editing of multiple genes. Altogether, our results considerably expand the Base Editor application to introduce human disease-causing mutations in zebrafish. Base Editors are emerging as an innovative technology to introduce point mutations in complex genomes. Here the authors describe a near PAM-less base editor and its application in zebrafish to efficiently create disease models harbouring specific point mutations.
Collapse
|
47
|
Mytlis A, Kumar V, Qiu T, Deis R, Hart N, Levy K, Masek M, Shawahny A, Ahmad A, Eitan H, Nather F, Adar-Levor S, Birnbaum RY, Elia N, Bachmann-Gagescu R, Roy S, Elkouby YM. Control of meiotic chromosomal bouquet and germ cell morphogenesis by the zygotene cilium. Science 2022; 376:eabh3104. [PMID: 35549308 DOI: 10.1126/science.abh3104] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A hallmark of meiosis is chromosomal pairing, which requires telomere tethering and rotation on the nuclear envelope via microtubules, driving chromosome homology searches. Telomere pulling toward the centrosome forms the "zygotene chromosomal bouquet". Here, we identified the "zygotene cilium" in oocytes. This cilium provides a cable system for the bouquet machinery, extending throughout the germline cyst. Using zebrafish mutants and live manipulations, we demonstrate that the cilium anchors the centrosome to counterbalance telomere pulling. The cilium is essential for bouquet and synaptonemal complex formation, oogenesis, ovarian development, and fertility. Thus, a cilium represents a conserved player in zebrafish and mouse meiosis, which sheds light on reproductive aspects in ciliopathies, and suggests that cilia can control chromosomal dynamics.
Collapse
Affiliation(s)
- Avishag Mytlis
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Vineet Kumar
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Tao Qiu
- Institute of Molecular and Cell Biology, Proteos, 138673 Singapore
| | - Rachael Deis
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Neta Hart
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Karine Levy
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Markus Masek
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland.,Institute of Medical Genetics, University of Zurich, 8952 Schlieren, Switzerland
| | - Amal Shawahny
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Adam Ahmad
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Hagai Eitan
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Farouq Nather
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Shai Adar-Levor
- Departments of Life Sciences, Ben-Gurion University of the Negev, Beer Shave 84105, Israel
| | - Ramon Y Birnbaum
- Departments of Life Sciences, Ben-Gurion University of the Negev, Beer Shave 84105, Israel
| | - Natalie Elia
- Departments of Life Sciences, Ben-Gurion University of the Negev, Beer Shave 84105, Israel
| | - Ruxandra Bachmann-Gagescu
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland.,Institute of Medical Genetics, University of Zurich, 8952 Schlieren, Switzerland
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Proteos, 138673 Singapore.,Department of Biological Sciences, National University of Singapore, 117543 Singapore.,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 119288 Singapore
| | - Yaniv M Elkouby
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| |
Collapse
|
48
|
Amanda S, Tan TK, Ong JZL, Theardy MS, Wong RWJ, Huang XZ, Ali MZ, Li Y, Gong Z, Inagaki H, Foo EY, Pang B, Tan SY, Iida S, Sanda T. IRF4 drives clonal evolution and lineage choice in a zebrafish model of T-cell lymphoma. Nat Commun 2022; 13:2420. [PMID: 35504924 PMCID: PMC9065160 DOI: 10.1038/s41467-022-30053-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
IRF4 is a master regulator of immunity and is also frequently overexpressed in mature lymphoid neoplasms. Here, we demonstrate the oncogenicity of IRF4 in vivo, its potential effects on T-cell development and clonal evolution using a zebrafish model. IRF4-transgenic zebrafish develop aggressive tumors with massive infiltration of abnormal lymphocytes that spread to distal organs. Many late-stage tumors are mono- or oligoclonal, and tumor cells can expand in recipient animals after transplantation, demonstrating their malignancy. Mutation of p53 accelerates tumor onset, increases penetrance, and results in tumor heterogeneity. Surprisingly, single-cell RNA-sequencing reveals that the majority of tumor cells are double-negative T-cells, many of which express tcr-γ that became dominant as the tumors progress, whereas double-positive T-cells are largely diminished. Gene expression and epigenetic profiling demonstrates that gata3, mycb, lrrn1, patl1 and psip1 are specifically activated in tumors, while genes responsible for T-cell differentiation including id3 are repressed. IRF4-driven tumors are sensitive to the BRD inhibitor.
Collapse
Affiliation(s)
- Stella Amanda
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Tze King Tan
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Jolynn Zu Lin Ong
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | | | - Regina Wan Ju Wong
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Muhammad Zulfaqar Ali
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Yan Li
- Department of Biological Sciences, National University of Singapore, 117543, Singapore, Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, 117543, Singapore, Singapore
| | - Hiroshi Inagaki
- Department of Pathology and Molecular Diagnostics, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Ee Yong Foo
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Brendan Pang
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Soo Yong Tan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore.
| |
Collapse
|
49
|
Yang S, Ning G, Hou Y, Cao Y, Xu J, Wu J, Zhang T, Wang Q. Myoneurin regulates BMP signaling by competing with Ppm1a for Smad binding. iScience 2022; 25:104495. [PMID: 35712083 PMCID: PMC9194458 DOI: 10.1016/j.isci.2022.104495] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 04/07/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022] Open
Abstract
A delicate balance of BMP activity is critical for tissue formation and organogenesis. However, the mechanical molecular details in ensuring the proper duration and intensity of BMP signaling have yet to be fully elucidated. Here, we identified a zebrafish mutant with a disrupted gene encoding for the BTB/POZ and zinc finger protein myoneurin (Mynn). mynn−/− mutants exhibited severe loss of pharyngeal cartilage elements, owing to poor proliferation, blocked differentiation, and low viability of cranial neural crest cells. Depletion of mynn in both zebrafish embryos and mammalian cells led to a reduction of the BMP signal activity. Mechanistically, Mynn interacts with Smad proteins in the nucleus, thereby disrupting the association between Smad protein and the phosphatase Ppm1a. Ultimately, this interaction prevents Smad dephosphorylation. More broadly, our findings may provide a new strategy to balance BMP signal activity via competitive binding of Mynn and Ppm1a to Smad proteins during pharyngeal cartilage formation. mynn gene is essential for pharyngeal cartilage development mynn is required for the proliferation, differentiation, and survival of the CNCCs Mynn has an evolutionarily conserved function in supporting BMP signal Mynn maintains BMP signal activity by competing with Ppm1a for Smad binding
Collapse
|
50
|
Cascallar M, Alijas S, Pensado-López A, Vázquez-Ríos AJ, Sánchez L, Piñeiro R, de la Fuente M. What Zebrafish and Nanotechnology Can Offer for Cancer Treatments in the Age of Personalized Medicine. Cancers (Basel) 2022; 14:cancers14092238. [PMID: 35565373 PMCID: PMC9099873 DOI: 10.3390/cancers14092238] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer causes millions of deaths each year and thus urgently requires the development of new therapeutic strategies. Nanotechnology-based anticancer therapies are a promising approach, with several formulations already approved and in clinical use. The evaluation of these therapies requires efficient in vivo models to study their behavior and interaction with cancer cells, and to optimize their properties to ensure maximum efficacy and safety. In this way, zebrafish is an important candidate due to its high homology with the human genoma, its large offspring, and the ease in developing specific cancer models. The role of zebrafish as a model for anticancer therapy studies has been highly evidenced, allowing researchers not only to perform drug screenings but also to evaluate novel therapies such as immunotherapies and nanotherapies. Beyond that, zebrafish can be used as an “avatar” model for performing patient-derived xenografts for personalized medicine. These characteristics place zebrafish in an attractive position as a role model for evaluating novel therapies for cancer treatment, such as nanomedicine.
Collapse
Affiliation(s)
- María Cascallar
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Abi Judit Vázquez-Ríos
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- DIVERSA Technologies S.L., 15782 Santiago de Compostela, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Roberto Piñeiro
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- DIVERSA Technologies S.L., 15782 Santiago de Compostela, Spain
- Correspondence: ; Tel.: +34-981-955-704
| |
Collapse
|