Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Katherine A. Jones is active.

Publication


Featured researches published by Katherine A. Jones.


Cell | 1998

A Novel CDK9-Associated C-Type Cyclin Interacts Directly with HIV-1 Tat and Mediates Its High-Affinity, Loop-Specific Binding to TAR RNA

Ping Wei; Mitchell E Garber; Shi-Min Fang; Wolfgang H. Fischer; Katherine A. Jones

The HIV-1 Tat protein regulates transcription elongation through binding to the viral TAR RNA stem-loop structure. We have isolated a novel 87 kDa cyclin C-related protein (cyclin T) that interacts specifically with the transactivation domain of Tat. Cyclin T is a partner for CDK9, an RNAPII transcription elongation factor. Remarkably, the interaction of Tat with cyclin T strongly enhances the affinity and specificity of the Tat:TAR RNA interaction, and confers a requirement for sequences in the loop of TAR that are not recognized by Tat alone. Moreover, overexpression of human cyclin T rescues Tat activity in nonpermissive rodent cells. We propose that Tat directs cyclin T-CDK9 to RNAPII through cooperative binding to TAR RNA.


Molecular and Cellular Biology | 2000

CDK9 Autophosphorylation Regulates High-Affinity Binding of the Human Immunodeficiency Virus Type 1 Tat–P-TEFb Complex to TAR RNA

Mitchell E Garber; Timothy P. Mayall; Eric M. Suess; Jill Meisenhelder; Nancy Thompson; Katherine A. Jones

ABSTRACT Human immunodeficiency virus type 1 (HIV-1) Tat interacts with cyclin T1 (CycT1), a regulatory partner of CDK9 in the positive transcription elongation factor (P-TEFb) complex, and binds cooperatively with CycT1 to TAR RNA to recruit P-TEFb and promote transcription elongation. We show here that Tat also stimulates phosphorylation of affinity-purified core RNA polymerase II and glutathioneS-transferase–C-terminal-domain substrates by CycT1-CDK9, but not CycH-CDK7, in vitro. Interestingly, incubation of recombinant Tat–P-TEFb complexes with ATP enhanced binding to TAR RNA dramatically, and the C-terminal half of CycT1 masked binding of Tat to TAR RNA in the absence of ATP. ATP incubation lead to autophosphorylation of CDK9 at multiple C-terminal Ser and Thr residues, and full-length CycT1 (amino acids 728) [CycT1(1–728)], but not truncated CycT1(1–303), was also phosphorylated by CDK9. P-TEFb complexes containing a catalytically inactive CDK9 mutant (D167N) bound TAR RNA weakly and independently of ATP, as did a C-terminal truncated CDK9 mutant that was catalytically active but unable to undergo autophosphorylation. Analysis of different Tat proteins revealed that the 101-amino-acid SF2 HIV-1 Tat was unable to bind TAR with CycT1(1–303) in the absence of phosphorylated CDK9, whereas unphosphorylated CDK9 strongly blocked binding of HIV-2 Tat to TAR RNA in a manner that was reversed upon autophosphorylation. Replacement of CDK9 phosphorylation sites with negatively charged residues restored binding of CycT1(1–303)-D167N-Tat, and rendered D167N a more potent inhibitor of transcription in vitro. Taken together, these results demonstrate that CDK9 phosphorylation is required for high-affinity binding of Tat–P-TEFb to TAR RNA and that the state of P-TEFb phosphorylation may regulate Tat transactivation in vivo.


Current Opinion in Immunology | 1999

HIV-1 Tat: coping with negative elongation factors

Mitchell Garber; Katherine A. Jones

The intrinsic processivity of RNA polymerase II complexes arises from a complex interplay between the recently identified positive transcription elongation factor b (P-TEFb) and negative transcription elongation factors, DSIF (5, 6-dichloro-1-beta-D-ribofuranosylbenzimidazole [DRB]-sensitivity-inducing factor) and the negative elongation factor complex (NELF). Elements in nascent HIV-1 RNA function in concert with these factors and the HIV-1 Tat protein to ensure that viral transcription is induced strongly in activated T cells. Studies in the past year have elucidated key aspects of the Tat trans-activation mechanism that help to define this important paradigm for RNA-mediated control of transcription elongation.


Current Biology | 2006

CK2 Controls the Recruitment of Wnt Regulators to Target Genes In Vivo

Song Wang; Katherine A. Jones

Nuclear beta-catenin is a transcriptional coactivator of LEF-1/TCF DNA-binding proteins in the Wnt/Wg signaling pathway. Casein Kinase 2 (CK2), a positive regulator of Wnt signaling, is present in beta-catenin complexes and activated in Wnt-signaling cells. We show here that CK2 enhances beta-catenin:LEF-1 transactivation in vivo and in vitro and that beta-catenin and CK2 cycle on and off the DNA in an alternating manner with the TLE1 corepressor at Wnt target genes. Interestingly, CK2 phosphorylates hLEF-1 directly and stimulates binding and transactivation of beta-catenin:LEF-1 complexes on chromatin templates in vitro. In vitro, CK2 phosphorylation of hLEF-1 strongly enhances its affinity for beta-catenin and reduces its affinity for TLE1. MALDI-TOF mass spectrometry (MS) identified two CK2 phosphorylation sites (S42, S61) within the amino terminus of hLEF-1, and mutation of these sites reduced binding to beta-catenin in vitro and transactivation in vivo. Remarkably, treatment of cells with TBB, a pharmaceutical inhibitor of CK2, blocked the recruitment and cycling of beta-catenin and TLE1 at Wnt target genes in vivo. Taken together, these data indicate that CK2 is required for the assembly and cycling of Wnt-enhancer complexes in vivo.


Current Opinion in Cell Biology | 1993

Tat and the HIV-1 promoter.

Katherine A. Jones

The HIV-1 Tat protein enhances the formation of productive RNA polymerase II elongation complexes, potentially acting through a positive-acting, DRB-sensitive elongation factor. Tat is usually recruited to the HIV-1 promoter through the Tat trans-activation response element RNA stem-loop structure; however, recent data suggest that in certain cell types it can be directed instead through upstream enhancer elements. New studies also reveal that the response element overlaps a novel motif that promotes the assembly of abortive elongation complexes in the absence of Tat.


Genes & Development | 2013

α-Catenin interacts with APC to regulate β-catenin proteolysis and transcriptional repression of Wnt target genes

Seung H. Choi; Conchi Estarás; James J. Moresco; John R. Yates; Katherine A. Jones

Mutation of the adenomatous polyposis coli (APC) tumor suppressor stabilizes β-catenin and aberrantly reactivates Wnt/β-catenin target genes in colon cancer. APC mutants in cancer frequently lack the conserved catenin inhibitory domain (CID), which is essential for β-catenin proteolysis. Here we show that the APC CID interacts with α-catenin, a Hippo signaling regulator and heterodimeric partner of β-catenin at cell:cell adherens junctions. Importantly, α-catenin promotes β-catenin ubiquitylation and proteolysis by stabilizing its association with APC and protecting the phosphodegron. Moreover, β-catenin ubiquitylation requires binding to α-catenin. Multidimensional protein identification technology (MudPIT) proteomics of multiple Wnt regulatory complexes reveals that α-catenin binds with β-catenin to LEF-1/TCF DNA-binding proteins in Wnt3a signaling cells and recruits APC in a complex with the CtBP:CoREST:LSD1 histone H3K4 demethylase to regulate transcription and β-catenin occupancy at Wnt target genes. Interestingly, tyrosine phosphorylation of α-catenin at Y177 disrupts binding to APC but not β-catenin and prevents repression of Wnt target genes in transformed cells. Chromatin immunoprecipitation studies further show that α-catenin and APC are recruited with β-catenin to Wnt response elements in human embryonic stem cells (hESCs). Knockdown of α-catenin in hESCs prevents the switch-off of Wnt/β-catenin transcription and promotes endodermal differentiation. Our findings indicate a role for α-catenin in the APC destruction complex and at Wnt target genes.


Molecular Cell | 2009

SKIP interacts with c-Myc and Menin to promote HIV-1 Tat Transactivation

Vanessa Brès; Tomonori Yoshida; Loni Pickle; Katherine A. Jones

The Ski-interacting protein SKIP/SNW1 associates with the P-TEFb/CDK9 elongation factor and coactivates inducible genes, including HIV-1. We show here that SKIP also associates with c-Myc and Menin, a subunit of the MLL1 histone methyltransferase (H3K4me3) complex and that HIV-1 Tat transactivation requires c-Myc and Menin, but not MLL1 or H3K4me3. RNAi-ChIP experiments reveal that SKIP acts downstream of Tat:P-TEFb to recruit c-Myc and its partner TRRAP, a scaffold for histone acetyltransferases, to the HIV-1 promoter. By contrast, SKIP is recruited by the RNF20 H2B ubiquitin ligase to the basal HIV-1 promoter in a step that is bypassed by Tat and downregulated by c-Myc. Of interest, we find that SKIP and P-TEFb are dispensable for UV stress-induced HIV-1 transcription, which is strongly upregulated by treating cells with the CDK9 inhibitor flavopiridol. Thus, SKIP acts with c-Myc and Menin to promote HIV-1 Tat:P-TEFb transcription at an elongation step that is bypassed under stress.


Molecular Cell | 2015

SMADs and YAP Compete to Control Elongation of β-Catenin:LEF-1-Recruited RNAPII during hESC Differentiation

Conchi Estarás; Christopher Benner; Katherine A. Jones

The Wnt3a/β-catenin and Activin/SMAD2,3 signaling pathways synergize to induce endodermal differentiation of human embryonic stem cells; however, the underlying mechanism is not well understood. Using ChIP-seq and GRO-seq analyses, we show here that Wnt3a-induced β-catenin:LEF-1 enhancers recruit cohesin to direct enhancer-promoter looping and activate mesendodermal (ME) lineage genes. Moreover, we find that LEF-1 and other hESC enhancers recruit RNAPII complexes (eRNAPII) that are highly phosphorylated at Ser5, but not Ser7. Wnt3a signaling further increases Ser5P-RNAPII at LEF-1 sites and ME gene promoters, indicating that elongation remains limiting. However, subsequent Activin/SMAD2,3 signaling selectively increases transcription elongation, P-TEFb occupancy, and Ser7P-RNAPII levels at these genes. Finally, we show that the Hippo regulator, YAP, functions with TEAD to regulate binding of the NELF negative elongation factor and block SMAD2,3 induction of ME genes. Thus, the Wnt3a/β-catenin and Activin/SMAD2,3 pathways act in concert to counteract YAP repression and upregulate ME genes during early hESC differentiation.


Epigenetics | 2013

The cellular ratio of immune tolerance (immunoCRIT) is a definite marker for aggressiveness of solid tumors and may explain tumor dissemination patterns

Ivana Türbachova; Tim Schwachula; Ines Vasconcelos; Alexander Mustea; Tina Baldinger; Katherine A. Jones; Hermann Bujard; Alexander Olek; K. Olek; Katharina Gellhaus; Ioana Braicu; Dominique Könsgen; Christy Fryer; Elisabetta Ravot; Alexander Hellwag; Nicole Westerfeld; Oliver J. Gruss; Markus Meissner; Mazahir T. Hasan; Michael Weber; Ulrich Hoffmüller; Sven Zimmermann; Christoph Loddenkemper; Sven Mahner; Nina Babel; Els M. J. J. Berns; Richard Adams; Robert Zeilinger; Udo Baron; Ignace Vergote

The adaptive immune system is involved in tumor establishment and aggressiveness. Tumors of the ovaries, an immune-privileged organ, spread via transceolomic routes and rarely to distant organs. This is contrary to tumors of non-immune privileged organs, which often disseminate hematogenously to distant organs. Epigenetics-based immune cell quantification allows direct comparison of the immune status in benign and malignant tissues and in blood. Here, we introduce the “cellular ratio of immune tolerance” (immunoCRIT) as defined by the ratio of regulatory T cells to total T lymphocytes. The immunoCRIT was analyzed on 273 benign tissue samples of colorectal, bronchial, renal and ovarian origin as well as in 808 samples from primary colorectal, bronchial, mammary and ovarian cancers. ImmunoCRIT is strongly increased in all cancerous tissues and gradually augmented strictly dependent on tumor aggressiveness. In peripheral blood of ovarian cancer patients, immunoCRIT incrementally increases from primary diagnosis to disease recurrence, at which distant metastases frequently occur. We postulate that non-pathological immunoCRIT values observed in peripheral blood of immune privileged ovarian tumor patients are sufficient to prevent hematogenous spread at primary diagnosis. Contrarily, non-immune privileged tumors establish high immunoCRIT in an immunological environment equivalent to the bloodstream and thus spread hematogenously to distant organs. In summary, our data suggest that the immunoCRIT is a powerful marker for tumor aggressiveness and disease dissemination.


Seminars in Virology | 1993

Tat and the HIV-1 promoter: A model for RNA-mediated regulation of transcription

Philip L. Sheridan; Christian T. Sheline; Lawrence H. Milocco; Katherine A. Jones

The human immunodeficiency virus (HIV-1) promoter is unique among RNA pol II transcription units in that it contains an element in the leader RNA (the trans-activation response element, or TAR) which regulates transcriptional induction by the virus-encoded regulatory protein, Tat. Tat acts through the TAR element to increase the efficiency of RNA transcript elongation, and under selected circumstances is also capable of enhancing the RNA initiation rate. Transcripts that initiate in the absence of Tat are elongated inefficiently and terminate at random, a process that we will refer to as attenuation. In this review, we consider the mechanism of trans-activation by Tat (including the role of TAR RNA) as well as the influence of host cell DNAbinding proteins on the assembly of productive and abortive transcription complexes at the HIV-1 promoter.

Collaboration


Dive into the Katherine A. Jones's collaboration.

Top Co-Authors

Avatar

Philip L. Sheridan

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Ping Wei

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Mitchell E Garber

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Vanessa Brès

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Conchi Estarás

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Seung H. Choi

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Andrew P. Rice

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Christy J. Fryer

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

James J. Moresco

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge