Cargando…

CRISPR/CAS9-mediated knockout of Abi1 inhibits p185(Bcr-Abl)-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways

BACKGROUND: Abl interactor 1 (Abi1) is a downstream target of Abl tyrosine kinases and a component of the WAVE regulatory complex (WRC) that plays an important role in regulating actin cytoskeleton remodeling and membrane receptor signaling. While studies using short hairpin RNA (shRNA) have suggest...

Descripción completa

Detalles Bibliográficos
Autores principales: Faulkner, James, Jiang, Peixin, Farris, Delaney, Walker, Ryan, Dai, Zonghan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7147029/
https://www.ncbi.nlm.nih.gov/pubmed/32276588
http://dx.doi.org/10.1186/s13045-020-00867-5
_version_ 1783520337023991808
author Faulkner, James
Jiang, Peixin
Farris, Delaney
Walker, Ryan
Dai, Zonghan
author_facet Faulkner, James
Jiang, Peixin
Farris, Delaney
Walker, Ryan
Dai, Zonghan
author_sort Faulkner, James
collection PubMed
description BACKGROUND: Abl interactor 1 (Abi1) is a downstream target of Abl tyrosine kinases and a component of the WAVE regulatory complex (WRC) that plays an important role in regulating actin cytoskeleton remodeling and membrane receptor signaling. While studies using short hairpin RNA (shRNA) have suggested that Abi1 plays a critical role in Bcr-Abl-induced leukemogenesis, the mechanism involved is not clear. METHODS: In this study, we knocked out Abi1 expression in p185(Bcr-Abl)-transformed hematopoietic cells using CRISPR/Cas9-mediated gene editing technology. The effects of Abi1 deficiency on actin cytoskeleton remodeling, the Bcr-Abl signaling, IL-3 independent growth, and SDF-induced chemotaxis in these cells were examined by various in vitro assays. The leukemogenic activity of these cells was evaluated by a syngeneic mouse transplantation model. RESULTS: We show here that Abi1 deficiency reduced the IL3-independent growth and SDF-1α-mediated chemotaxis in p185(Bcr-Abl)-transformed hematopoietic cells and inhibited Bcr-Abl-induced abnormal actin remodeling. Depletion of Abi1 also impaired the Bcr-Abl signaling to the ERK and PI3 kinase/Akt pathways. Remarkably, the p185(Bcr-Abl)-transformed cells with Abi1 deficiency lost their ability to develop leukemia in syngeneic mice. Even though these cells developed drug tolerance in vitro after prolonged selection with imatinib as their parental cells, the imatinib-tolerant cells remain incapable of leukemogenesis in vivo. CONCLUSIONS: Together, this study highlights an essential role of Abi1 in Bcr-Abl-induced leukemogenesis and provides a model system for dissecting the Abi1 signaling in Bcr-Abl-positive leukemia.
format Online
Article
Text
id pubmed-7147029
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-71470292020-04-18 CRISPR/CAS9-mediated knockout of Abi1 inhibits p185(Bcr-Abl)-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways Faulkner, James Jiang, Peixin Farris, Delaney Walker, Ryan Dai, Zonghan J Hematol Oncol Research BACKGROUND: Abl interactor 1 (Abi1) is a downstream target of Abl tyrosine kinases and a component of the WAVE regulatory complex (WRC) that plays an important role in regulating actin cytoskeleton remodeling and membrane receptor signaling. While studies using short hairpin RNA (shRNA) have suggested that Abi1 plays a critical role in Bcr-Abl-induced leukemogenesis, the mechanism involved is not clear. METHODS: In this study, we knocked out Abi1 expression in p185(Bcr-Abl)-transformed hematopoietic cells using CRISPR/Cas9-mediated gene editing technology. The effects of Abi1 deficiency on actin cytoskeleton remodeling, the Bcr-Abl signaling, IL-3 independent growth, and SDF-induced chemotaxis in these cells were examined by various in vitro assays. The leukemogenic activity of these cells was evaluated by a syngeneic mouse transplantation model. RESULTS: We show here that Abi1 deficiency reduced the IL3-independent growth and SDF-1α-mediated chemotaxis in p185(Bcr-Abl)-transformed hematopoietic cells and inhibited Bcr-Abl-induced abnormal actin remodeling. Depletion of Abi1 also impaired the Bcr-Abl signaling to the ERK and PI3 kinase/Akt pathways. Remarkably, the p185(Bcr-Abl)-transformed cells with Abi1 deficiency lost their ability to develop leukemia in syngeneic mice. Even though these cells developed drug tolerance in vitro after prolonged selection with imatinib as their parental cells, the imatinib-tolerant cells remain incapable of leukemogenesis in vivo. CONCLUSIONS: Together, this study highlights an essential role of Abi1 in Bcr-Abl-induced leukemogenesis and provides a model system for dissecting the Abi1 signaling in Bcr-Abl-positive leukemia. BioMed Central 2020-04-10 /pmc/articles/PMC7147029/ /pubmed/32276588 http://dx.doi.org/10.1186/s13045-020-00867-5 Text en © The Author(s) 2020 Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Faulkner, James
Jiang, Peixin
Farris, Delaney
Walker, Ryan
Dai, Zonghan
CRISPR/CAS9-mediated knockout of Abi1 inhibits p185(Bcr-Abl)-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways
title CRISPR/CAS9-mediated knockout of Abi1 inhibits p185(Bcr-Abl)-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways
title_full CRISPR/CAS9-mediated knockout of Abi1 inhibits p185(Bcr-Abl)-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways
title_fullStr CRISPR/CAS9-mediated knockout of Abi1 inhibits p185(Bcr-Abl)-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways
title_full_unstemmed CRISPR/CAS9-mediated knockout of Abi1 inhibits p185(Bcr-Abl)-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways
title_short CRISPR/CAS9-mediated knockout of Abi1 inhibits p185(Bcr-Abl)-induced leukemogenesis and signal transduction to ERK and PI3K/Akt pathways
title_sort crispr/cas9-mediated knockout of abi1 inhibits p185(bcr-abl)-induced leukemogenesis and signal transduction to erk and pi3k/akt pathways
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7147029/
https://www.ncbi.nlm.nih.gov/pubmed/32276588
http://dx.doi.org/10.1186/s13045-020-00867-5
work_keys_str_mv AT faulknerjames crisprcas9mediatedknockoutofabi1inhibitsp185bcrablinducedleukemogenesisandsignaltransductiontoerkandpi3kaktpathways
AT jiangpeixin crisprcas9mediatedknockoutofabi1inhibitsp185bcrablinducedleukemogenesisandsignaltransductiontoerkandpi3kaktpathways
AT farrisdelaney crisprcas9mediatedknockoutofabi1inhibitsp185bcrablinducedleukemogenesisandsignaltransductiontoerkandpi3kaktpathways
AT walkerryan crisprcas9mediatedknockoutofabi1inhibitsp185bcrablinducedleukemogenesisandsignaltransductiontoerkandpi3kaktpathways
AT daizonghan crisprcas9mediatedknockoutofabi1inhibitsp185bcrablinducedleukemogenesisandsignaltransductiontoerkandpi3kaktpathways