Welcome to the new version of CaltechAUTHORS. Login is currently restricted to library staff. If you notice any issues, please email coda@library.caltech.edu
Published July 27, 2018 | Supplemental Material + Accepted Version
Journal Article Open

Imaging dynamic and selective low-complexity domain interactions that control gene transcription

Abstract

INTRODUCTION. DNA binding transcription factors (TFs) are quintessential regulators of eukaryotic gene expression. Early studies of TFs revealed their well-structured DNA binding domains (DBDs) and identified functionally critical activation domains (ADs) required for transcription. It later became evident that many ADs contain intrinsically disordered low-complexity sequence domains (LCDs), but how LCDs activate transcription has remained unclear. Although it is known that transcriptional activation by LCDs requires selective interaction with binding partners, it has been challenging to directly measure selective LCD-LCD recognition in vivo and unravel its mechanism of action. RATIONALE. Traditional biochemical reconstitution and genetics studies have identified most of the molecular players central to transcription regulation. However, the mechanism by which weak, dynamic protein-protein interactions drive gene activation in living cells has remained unknown. Advances in live-cell single-molecule imaging have opened a new frontier for studying transcription in vivo. In this study, we used synthetic LacO (Lac operator) arrays as well as endogenous GGAA microsatellite loci to study LCD-LCD interactions of TFs such as EWS/FLI1, TAF15, and Sp1 in live cells. To probe the dynamic behavior of TF LCDs at target genomic loci, we have combined CRISPR-Cas9 genome editing, mutagenesis, gene activation, cell transformation assays, and various high-resolution imaging approaches including fluorescence correlation spectroscopy, fluorescence recovery after photobleaching, lattice light-sheet microscopy, three-dimensional DNA fluorescence in situ hybridization, and live-cell single-particle tracking. RESULTS. Live-cell single-molecule imaging revealed that TF LCDs interact to form local high-concentration hubs at both synthetic DNA arrays and endogenous genomic loci. TF LCD hubs stabilize DNA binding, recruit RNA polymerase II (RNA Pol II), and activate transcription. LCD-LCD interactions within hubs are highly dynamic (seconds to minutes), selective for binding partners, and differentially sensitive to disruption by hexanediols. These findings suggest that under physiological conditions, rapid, reversible, and selective multivalent LCD-LCD interactions occur between TFs and the RNA Pol II machinery to activate transcription. We observed formation of functional TF LCD hubs at a wide range of intranuclear TF concentrations. Although we detected apparent liquid-liquid phase separation with gross overexpression of LCDs, transcriptionally competent TF LCD hubs were observed at physiological TF levels at endogenous chromosomal loci in the absence of detectable phase separation. In addition, mutagenesis, gene expression, and cell transformation assays in Ewing's sarcoma cells revealed a functional link between LCD-LCD interactions, transactivation capacity, and oncogenic potential. CONCLUSION. The use of various imaging methods in live cells powerfully complements in vitro studies and provides new insights into the nature of LCD interactions and their role in gene regulation. We propose that transactivation domains function by forming local high-concentration hubs of TFs via dynamic, multivalent, and specific LCD-LCD interactions. It also seems likely that weak, dynamic, and transient contacts between TFs play a role in disease-causing dysregulation of gene expression (i.e., EWS/FLI1 in Ewing's sarcoma), suggesting that LCD-LCD interactions may represent a new class of viable drug targets. Although we examined a small subset of TF LCDs, the principles uncovered regarding the dynamics and mechanisms driving LCD-LCD interactions may be applicable to other classes of proteins and biomolecular interactions occurring in many cell types.

Additional Information

© 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. 23 October 2017; resubmitted 12 March 2018. Accepted 13 June 2018. Published online 21 June 2018. We thank S. Lessnick, S. McKnight, and M. Kato for discussions and providing reagents; Q. Gan and A. Hansen for providing codes to analyze imaging data; J. Bosco and P. Sharma for help with molecular cloning; K. Heydari and the CRL Flow Cytometry Facility for assistance with flow cytometry; the CRL Molecular Imaging Center for providing access to confocal fluorescence microscopes; and J. Goodrich, G. S. Martin, and members of Tjian and Darzacq labs for critical reading of the manuscript. This work was supported by California Institute of Regenerative Medicine grant LA1-08013 (to X.D.), NIH grants UO1-EB021236 and U54-DK107980 (to X.D.), and the Howard Hughes Medical Institute (to Z.L., L.L., and R.T.). Author contributions: R.T. conceived of and supervised the project. S.C., R.T., and X.D. designed experiments and interpreted the data. S.C. performed genome editing, soft colony formation assays, live-cell imaging in the presence of hexanediols, confocal imaging, DNA FISH, immunofluorescence, FRAP and SPT measurements, and all of the imaging data analyses. C.D.-D. established and characterized Halo-RPB1 cell lines, characterized LacO-containing cell lines, and performed all of the RT-qPCR assays and related data analyses. S.C. and Z.L. developed codes for SPT data analyses. P.D. performed lattice light-sheet imaging. G.M.D. and S.C. designed and generated constructs. S.C. and C.C. performed luciferase assays. A.H. performed FCS measurements. S.B. and L.L. synthesized and characterized fluorescent HaloTag and SNAPf-tag ligands. S.C., R.T., C.D.-D., C.C., Z.L., X.D., P.D., and A.H. prepared the manuscript and supplementary materials. Competing interests: L.L. has filed a patent and patent applications (e.g., U.S. Patent 9,933,417) covering azetidine-containing fluorophores such as JF549. Data and materials availability: All codes for imaging data analyses have been submitted to GitHub and are publicly available (https://github.com/Shasha-Chong/CodeFor2018SciencePaper).

Attached Files

Accepted Version - nihms-1059185.pdf

Supplemental Material - aar2555_chong_sm.pdf

Files

nihms-1059185.pdf
Files (18.3 MB)
Name Size Download all
md5:d1edb2c9565fb3ca6bd6285e5d9938b5
2.6 MB Preview Download
md5:2099c9321bc2d217f296598304d99efd
15.7 MB Preview Download

Additional details

Created:
August 19, 2023
Modified:
October 23, 2023