Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
J Cell Sci ; 136(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-36995025

ABSTRACT

Switching genes on and off on cue is a cornerstone for understanding gene functions. One contemporary approach for loss-of-function studies of essential genes involves CRISPR-mediated knockout of the endogenous locus in conjunction with the expression of a rescue construct, which can subsequently be turned off to produce a gene inactivation effect in mammalian cell lines. A broadening of this approach would involve simultaneously switching on a second construct to interrogate the functions of a gene in the pathway. In this study, we developed a pair of switches that were independently controlled by both inducible promoters and degrons, enabling the toggling between two constructs with comparable kinetics and tightness. The gene-OFF switch was based on TRE transcriptional control coupled with auxin-induced degron-mediated proteolysis. A second independently controlled gene-ON switch was based on a modified ecdysone promoter and mutated FKBP12-derived destabilization domain degron, allowing acute and tuneable gene activation. This platform facilitates efficient generation of knockout cell lines containing a two-gene switch that is regulated tightly and can be flipped within a fraction of the time of a cell cycle.


Subject(s)
Gene Expression Regulation , Indoleacetic Acids , Animals , Cell Line , Indoleacetic Acids/pharmacology , Proteolysis , Promoter Regions, Genetic/genetics , Mammals/metabolism
2.
Cell Death Dis ; 15(1): 2, 2024 01 03.
Article in English | MEDLINE | ID: mdl-38172496

ABSTRACT

Mitotic catastrophe induced by prolonged mitotic arrest is a major anticancer strategy. Although antiapoptotic BCL2-like proteins, including BCL-XL, are known to regulate apoptosis during mitotic arrest, adaptive changes in their expression can complicate loss-of-function studies. Our studies revealed compensatory alterations in the expression of BCL2 and MCL1 when BCL-XL is either downregulated or overexpressed. To circumvent their reciprocal regulation, we utilized a degron-mediated system to acutely silence BCL-XL just before mitosis. Our results show that in epithelial cell lines including HeLa and RPE1, BCL-XL and BCL2 acted collaboratively to suppress apoptosis during both unperturbed cell cycle and mitotic arrest. By tagging BCL-XL and BCL2 with a common epitope, we estimated that BCL-XL was less abundant than BCL2 in the cell. Nonetheless, BCL-XL played a more prominent antiapoptotic function than BCL2 during interphase and mitotic arrest. Loss of BCL-XL led to mitotic cell death primarily through a BAX-dependent process. Furthermore, silencing of BCL-XL led to the stabilization of MCL1, which played a significant role in buffering apoptosis during mitotic arrest. Nevertheless, even in a MCL1-deficient background, depletion of BCL-XL accelerated mitotic apoptosis. These findings underscore the pivotal involvement of BCL-XL in controlling timely apoptosis during mitotic arrest, despite adaptive changes in the expression of other BCL2-like proteins.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2 , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism , Cell Line, Tumor , Apoptosis/genetics
3.
Mol Biol Cell ; 32(14): 1320-1330, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33979199

ABSTRACT

Loss-of-function analysis is one of the major arsenals we have for understanding gene functions in mammalian cells. For analysis of essential genes, the major challenge is to develop simple methodologies for tight and rapid inducible gene inactivation. One approach involves CRISPR-Cas9-mediated disruption of the endogenous locus in conjunction with the expression of a rescue construct, which can subsequently be turned off to produce a gene inactivation effect. Here we describe the development of a set of Sleeping Beauty transposon-based vectors for expressing auxin-inducible degron (AID)-tagged genes under the regulation of a tetracycline-controlled promoter. The dual transcriptional and degron-mediated post-translational regulation allows rapid and tight silencing of protein expression in mammalian cells. We demonstrated that both non-essential and essential genes could be targeted in human cell lines using a one-step transfection method. Moreover, multiple genes could be simultaneously or sequentially targeted, allowing inducible inactivation of multiple genes. These resources enable highly efficient generation of conditional gene silencing cell lines to facilitate functional studies of essential genes.


Subject(s)
Gene Expression Regulation/genetics , Gene Silencing/physiology , Genetic Engineering/methods , Animals , CRISPR-Cas Systems , Cell Line , Gene Expression/genetics , Gene Expression Regulation/physiology , Genes, Essential/genetics , Genes, Reporter/genetics , Genetic Vectors/genetics , Humans , Indoleacetic Acids/metabolism , Loss of Function Mutation/genetics , Promoter Regions, Genetic/genetics , Transfection , Transposases/genetics , Transposases/metabolism
4.
Cell Rep ; 37(2): 109808, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34644583

ABSTRACT

One of the most intriguing features of cell-cycle control is that, although there are multiple cyclin-dependent kinases (CDKs) in higher eukaryotes, a single CDK is responsible for both G1-S and G2-M in yeasts. By leveraging a rapid conditional silencing system in human cell lines, we confirm that CDK1 assumes the role of G1-S CDK in the absence of CDK2. Unexpectedly, CDK1 deficiency does not prevent mitotic entry. Nonetheless, inadequate phosphorylation of mitotic substrates by noncanonical cyclin B-CDK2 complexes does not allow progression beyond metaphase and underscores deleterious late mitotic events, including the uncoupling of anaphase A and B and cytokinesis. Elevation of CDK2 to a level similar to CDK1 overcomes the mitotic defects caused by CDK1 deficiency, indicating that the relatively low concentration of CDK2 accounts for the defective anaphase. Collectively, these results reveal that the difference between G2-M and G1-S CDKs in human cells is essentially quantitative.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Proliferation , Mitosis , Retinal Pigment Epithelium/enzymology , Uterine Cervical Neoplasms/enzymology , CDC2 Protein Kinase/genetics , Cyclin B/genetics , Cyclin B/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Phosphorylation , Signal Transduction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL