scholarly journals Cdc37 engages in stable, S14A mutation-reinforced association with the most atypical member of the yeast kinome, Cdk-activating kinase (Cak1)

Author(s):  
Stefan Millson ◽  
Patricija van Oosten-Hawle ◽  
Mohammed A. Alkuriji ◽  
Andrew Truman ◽  
Marco Siderius ◽  
...  
1994 ◽  
Vol 269 (8) ◽  
pp. 6140-6148
Author(s):  
F.J. Johannes ◽  
J. Prestle ◽  
S. Eis ◽  
P. Oberhagemann ◽  
K. Pfizenmaier

Cell Cycle ◽  
2010 ◽  
Vol 9 (4) ◽  
pp. 689-699 ◽  
Author(s):  
Sabine Paternot ◽  
Laurence Bockstaele ◽  
Xavier Bisteau ◽  
Hugues Kooken ◽  
Katia Coulonval ◽  
...  

2020 ◽  
Vol 117 (37) ◽  
pp. 22849-22857 ◽  
Author(s):  
Basil J. Greber ◽  
Juan M. Perez-Bertoldi ◽  
Kif Lim ◽  
Anthony T. Iavarone ◽  
Daniel B. Toso ◽  
...  

The human CDK-activating kinase (CAK), a complex composed of cyclin-dependent kinase (CDK) 7, cyclin H, and MAT1, is a critical regulator of transcription initiation and the cell cycle. It acts by phosphorylating the C-terminal heptapeptide repeat domain of the RNA polymerase II (Pol II) subunit RPB1, which is an important regulatory event in transcription initiation by Pol II, and it phosphorylates the regulatory T-loop of CDKs that control cell cycle progression. Here, we have determined the three-dimensional (3D) structure of the catalytic module of human CAK, revealing the structural basis of its assembly and providing insight into CDK7 activation in this context. The unique third component of the complex, MAT1, substantially extends the interaction interface between CDK7 and cyclin H, explaining its role as a CAK assembly factor, and it forms interactions with the CDK7 T-loop, which may contribute to enhancing CAK activity. We have also determined the structure of the CAK in complex with the covalently bound inhibitor THZ1 in order to provide insight into the binding of inhibitors at the CDK7 active site and to aid in the rational design of therapeutic compounds.


2010 ◽  
Vol 84 (16) ◽  
pp. 8342-8347 ◽  
Author(s):  
Krishnamurthy Konduru ◽  
Gerardo G. Kaplan

ABSTRACT Hepatitis A virus (HAV), an atypical member of the Picornaviridae, grows poorly in cell culture. To define determinants of HAV growth, we introduced a blasticidin (Bsd) resistance gene into the virus genome and selected variants that grew at high concentrations of Bsd. The mutants grew fast and had increased rates of RNA replication and translation but did not produce significantly higher virus yields. Nucleotide sequence analysis and reverse genetic studies revealed that a T6069G change resulting in a F42L amino acid substitution in the viral polymerase (3Dpol) was required for growth at high Bsd concentrations whereas a silent C7027T mutation enhanced the growth rate. Here, we identified a novel determinant(s) in 3Dpol that controls the kinetics of HAV growth.


Nature ◽  
1994 ◽  
Vol 371 (6494) ◽  
pp. 254-257 ◽  
Author(s):  
Tomi P. Mäkelä ◽  
Jean-Pierre Tassan ◽  
Erich A. Nigg ◽  
Séverine Frutiger ◽  
Graham J. Hughes ◽  
...  

2008 ◽  
Vol 22 (1) ◽  
pp. 91-104 ◽  
Author(s):  
Aurélia E. Lewis ◽  
Marte Rusten ◽  
Erling A. Hoivik ◽  
Elisabeth L. Vikse ◽  
Magnus L. Hansson ◽  
...  

Abstract The nuclear receptor steroidogenic factor-1 (SF1) is critical for development and function of steroidogenic tissues. Posttranslational modifications are known to influence the transcriptional capacity of SF1, and it was previously demonstrated that serine 203 is phosphorylated. In this paper we report that serine 203 is phosphorylated by a cyclin-dependent kinase 7 (CDK7)-mediated process. As part of the CDK-activating kinase complex, CDK7 is a component of the basal transcription factor TFIIH, and phosphorylation of SF1 as well as SF1-dependent transcription was clearly reduced in cells carrying a mutation that renders the CDK-activating kinase complex unable to interact with the TFIIH core. Coimmunoprecipitation analyses revealed that SF1 and CDK7 reside in the same complex, and kinase assays demonstrated that immunoprecipitated CDK7 and purified TFIIH phosphorylate SF1 in vitro. The CDK inhibitor roscovitine blocked phosphorylation of SF1, and an inactive form of CDK7 repressed the phosphorylation level and the transactivation capacity of SF1. Structural studies have identified phosphoinositides as potential ligands for SF1. Interestingly, we found that mutations designed to block phospholipid binding dramatically decreased the level of SF1 phosphorylation. Together our results suggest a connection between ligand occupation and phosphorylation and association with the basic transcriptional machinery, indicating an intricate regulation of SF1 transactivation.


Sign in / Sign up

Export Citation Format

Share Document