Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors

Tag: CRISPR/Cas9

CRISPR/Cas9-induced disruption of Bodo saltans paraflagellar rod-2 gene reveals its importance for cell survival

Developing transfection protocols for marine protists is an emerging field that will allow the functional characterization of protist genes and their roles in organism responses to the environment. We developed a CRISPR/Cas9 editing protocol for Bodo saltans, a free-living kinetoplastid with tolerance to both marine and freshwater conditions, and a close non-parasitic relative of trypanosomatids. Our results show that SaCas9/sgRNA ribonucleoprotein (RNP) complex-mediated disruption of the paraflagellar rod 2 gene (BsPFR2) was achieved using electroporation-mediated transfection. The use of CRISPR/Cas9 genome editing can increase the efficiency of targeted homologous recombination when a repair DNA template is provided. Based on sequence analysis, two mechanisms for repairing double-strand breaks (DSB) in B. saltans are active; homologous directed repair (HDR) utilizing an exogenous DNA template that carries an antibiotic resistance gene, and non-homologous end joining (NHEJ). However, HDR was only achieved when a single (vs. multiple) SaCas9 RNP complex was provided. Further, the biallelic knockout of BsPFR2 was detrimental for the cell, highlighting its essential role for cell survival because it facilitates the movement of food particles into the cytostome. Our Cas9/sgRNA RNP complex protocol provides a new tool for assessing gene functions in B. saltans, and perhaps similar protists with polycistronic transcription. This article is protected by copyright. All rights reserved.

Fatma Gomaa, Zhu-Hong Li, David J Beaudoin, Heba Alzan, Peter R Girguis, Roberto Docampo, Virginia P Edgcomb. Environ Microbiol. 2022 Jan 31. doi: 10.1111/1462-2920.15918.

p53 Hinders CRISPR/Cas9-Mediated Targeted Gene Disruption in Memory CD8 T Cells In Vivo

CRISPR/Cas9 technology has revolutionized rapid and reliable gene editing in cells. Although many cell types have been subjected to CRISPR/Cas9-mediated gene editing, there is no evidence of success in genetic alteration of Ag-experienced memory CD8 T cells. In this study, we show that CRISPR/Cas9-mediated gene editing in memory CD8 T cells precludes their proliferation after Ag re-encounter in vivo. This defect is mediated by the proapoptotic transcription factor p53, a sensor of DNA damage. Temporarily inhibiting p53 function offers a window of opportunity for the memory CD8 T cells to repair the DNA damage, facilitating robust recall responses on Ag re-encounter. We demonstrate this by functionally altering memory CD8 T cells using CRISPR/Cas9-mediated targeted gene disruption under the aegis of p53siRNA in the mouse model. Our approach thus adapts the CRISPR/Cas9 technology for memory CD8 T cells to undertake gene editing in vivo, for the first time, to our knowledge.

Samarchith P. KurupSteven J. MoiofferLecia L. Pewe and John T. Harty. J Immunol. 2020 Sep 4;ji2000654. doi: 10.4049/jimmunol.2000654.