Cell transplantation into immune compromised mice has transformed our understanding of human disease and has been used extensively to assess regeneration, stem cell self-renewal, and cancer. Despite the great utility of xenograft cell transplantation into immune compromised mice, mouse models are not amenable to large-scale studies due to high husbandry cost and do not facilitate direct visualization of engrafted cells. B contrast, zebrafish are inexpensive, can be reared in large numbers, and are amenable to large-scale chemical genetic approaches where compounds can be added directly to the water. Optically clear caspar-strain zebrafish lack pigmentation, facilitating the direct visualization of fluorescent cells in live adult animals. Moreover, cell transplantation utilizing genetically-identical, syngeneic zebrafish strains has validated that large-scale cell transplantation using hundreds of recipient animals is possible. Finally, zebrafish can be raised at 35C and thus a potential model for engraftment of human and mouse cells. Despite these clear advantages of the zebrafish model, the use of immune compromised zebrafish for xenograft cell transplantation has yet to be reported. Capitalizing on optically-clear caspar-strain zebrafish and recently developed gene inactivation using TALENs, we will develop zebrafish lines with similar mutations found in immune compromised mice that are commonly employed for xenograft cell transplantation.
Aim 1 will develop zebrafish lines that are deficient in forkhead box N l (foxnl/nude), recombination-activating gene 2 (rag2), Janus kinase 3 (jak3), DNA-dependent protein kinase (prkdc), and interieukin 2-receptor gamma (IL2rg).
Aim 2 will assess each model for the ability to engraft fluorescent-labeled normal and malignant cells from zebrafish, mouse, and human. Transplant engraftment will be assessed in single mutant fish and compound mutants for 1) rag2-/-, jak3-/-;2) rag2-/-, 112rga-/-, 112rgb-/-;3) prkdc-/-, jak3-/-;and 4) prkc-/-, IL2rga-/-, Il2rgb-/-. These experiments will allow comprehensive comparison of engraftment between zebrafish mutant lines.
Aim 3 will establish an organizational structure to rapidly and efficiently distribute viable mutant lines to the research community. The experiments outlined here will develop a much-needed resource for the community, facilitating the next generation of low-cost, high throughput cell transplantation models. Such broad reaching applications for immune compromised zebrafish spans the mission of many NIH institutes.

Public Health Relevance

Capitalizing on transparent zebrafish and recently developed gene inactivation using TALENs, we propose to develop immune compromised zebrafish for use in xenograft cell transplantation. These models will facilitate direct visualization of engrafted cells, are amenable to large-scale transplantation studies, and will allow chemical genetic approaches to uncover pathways associated with regeneration, self-renewal, and cancer.

Agency
National Institute of Health (NIH)
Institute
Office of The Director, National Institutes of Health (OD)
Type
Resource-Related Research Projects (R24)
Project #
5R24OD016761-02
Application #
8711587
Study Section
Special Emphasis Panel (ZOD1)
Program Officer
Mirochnitchenko, Oleg
Project Start
2013-08-01
Project End
2017-07-31
Budget Start
2014-08-01
Budget End
2015-07-31
Support Year
2
Fiscal Year
2014
Total Cost
Indirect Cost
Name
Massachusetts General Hospital
Department
Type
DUNS #
City
Boston
State
MA
Country
United States
Zip Code
02199
Hayes, Madeline N; McCarthy, Karin; Jin, Alexander et al. (2018) Vangl2/RhoA Signaling Pathway Regulates Stem Cell Self-Renewal Programs and Growth in Rhabdomyosarcoma. Cell Stem Cell 22:414-427.e6
Kasheta, Melissa; Painter, Corrie A; Moore, Finola E et al. (2017) Identification and characterization of T reg-like cells in zebrafish. J Exp Med 214:3519-3530
Melancon, E; Gomez De La Torre Canny, S; Sichel, S et al. (2017) Best practices for germ-free derivation and gnotobiotic zebrafish husbandry. Methods Cell Biol 138:61-100
Tenente, InĂªs M; Hayes, Madeline N; Ignatius, Myron S et al. (2017) Myogenic regulatory transcription factors regulate growth in rhabdomyosarcoma. Elife 6:
Ignatius, Myron S; Hayes, Madeline N; Lobbardi, Riadh et al. (2017) The NOTCH1/SNAIL1/MEF2C Pathway Regulates Growth and Self-Renewal in Embryonal Rhabdomyosarcoma. Cell Rep 19:2304-2318
Fazio, Maurizio; Avagyan, Serine; van Rooijen, Ellen et al. (2017) Efficient Transduction of Zebrafish Melanoma Cell Lines and Embryos Using Lentiviral Vectors. Zebrafish 14:379-382
Davison, James M; Lickwar, Colin R; Song, Lingyun et al. (2017) Microbiota regulate intestinal epithelial gene expression by suppressing the transcription factor Hepatocyte nuclear factor 4 alpha. Genome Res 27:1195-1206
Tang, Qin; Iyer, Sowmya; Lobbardi, Riadh et al. (2017) Dissecting hematopoietic and renal cell heterogeneity in adult zebrafish at single-cell resolution using RNA sequencing. J Exp Med 214:2875-2887
Lickwar, Colin R; Camp, J Gray; Weiser, Matthew et al. (2017) Genomic dissection of conserved transcriptional regulation in intestinal epithelial cells. PLoS Biol 15:e2002054
Moore, John C; Langenau, David M (2016) Allograft Cancer Cell Transplantation in Zebrafish. Adv Exp Med Biol 916:265-87

Showing the most recent 10 out of 21 publications