Network


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

Hotspot


Dive into the research topics where Ursula Jakob is active.

Publication


Featured researches published by Ursula Jakob.


Cell | 1999

Chaperone Activity with a Redox Switch

Ursula Jakob; Wilson B. Muse; Markus Eser; James C. A. Bardwell

Hsp33, a member of a newly discovered heat shock protein family, was found to be a very potent molecular chaperone. Hsp33 is distinguished from all other known molecular chaperones by its mode of functional regulation. Its activity is redox regulated. Hsp33 is a cytoplasmically localized protein with highly reactive cysteines that respond quickly to changes in the redox environment. Oxidizing conditions like H2O2 cause disulfide bonds to form in Hsp33, a process that leads to the activation of its chaperone function. In vitro and in vivo experiments suggest that Hsp33 protects cells from oxidants, leading us to conclude that we have found a protein family that plays an important role in the bacterial defense system toward oxidative stress.


Trends in Biochemical Sciences | 1994

Assisting spontaneity: the role of Hsp90 and small Hsps as molecular chaperones

Ursula Jakob; Johannes Buchner

Hsp90 and small Hsps are two abundant types of eukaryotic stress protein whose function has remained largely enigmatic. In the cell, Hsp90 exists in a complex (with other Hsps and prolyl isomerases) possibly implicated in interactions with non-native proteins. Recent biochemical analysis of both Hsp90 and small Hsps has revealed that they may act as ATP-independent molecular chaperones involved in protein folding and unfolding events.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Quantifying changes in the thiol redox proteome upon oxidative stress in vivo

Lars I. Leichert; Florian Gehrke; Harini V. Gudiseva; Thomas W. Blackwell; Marianne Ilbert; Angela K. Walker; John R. Strahler; Philip C. Andrews; Ursula Jakob

Antimicrobial levels of reactive oxygen species (ROS) are produced by the mammalian host defense to kill invading bacteria and limit bacterial colonization. One main in vivo target of ROS is the thiol group of proteins. We have developed a quantitative thiol trapping technique termed OxICAT to identify physiologically important target proteins of hydrogen peroxide (H2O2) and hypochlorite (NaOCl) stress in vivo. OxICAT allows the precise quantification of oxidative thiol modifications in hundreds of different proteins in a single experiment. It also identifies the affected proteins and defines their redox-sensitive cysteine(s). Using this technique, we identified a group of Escherichia coli proteins with significantly (30–90%) oxidatively modified thiol groups, which appear to be specifically sensitive to either H2O2 or NaOCl stress. These results indicate that individual oxidants target distinct proteins in vivo. Conditionally essential E. coli genes encode one-third of redox-sensitive proteins, a finding that might explain the bacteriostatic effect of oxidative stress treatment. We identified a select group of redox-regulated proteins, which protect E. coli against oxidative stress conditions. These experiments illustrate that OxICAT, which can be used in a variety of different cell types and organisms, is a powerful tool to identify, quantify, and monitor oxidative thiol modifications in vivo.


Cell | 2008

Bleach Activates A Redox-Regulated Chaperone by Oxidative Protein Unfolding

Jeannette Winter; Marianne Ilbert; Paul C.F. Graf; D. Özcelik; Ursula Jakob

Hypochlorous acid (HOCl), the active ingredient in household bleach, is an effective antimicrobial produced by the mammalian host defense to kill invading microorganisms. Despite the widespread use of HOCl, surprisingly little is known about its mode of action. In this study, we demonstrate that low molar ratios of HOCl to protein cause oxidative protein unfolding in vitro and target thermolabile proteins for irreversible aggregation in vivo. As a defense mechanism, bacteria use the redox-regulated chaperone Hsp33, which responds to bleach treatment with the reversible oxidative unfolding of its C-terminal redox switch domain. HOCl-mediated unfolding turns inactive Hsp33 into a highly active chaperone holdase, which protects essential Escherichia coli proteins against HOCl-induced aggregation and increases bacterial HOCl resistance. Our results substantially improve our molecular understanding about HOCls functional mechanism. They suggest that the antimicrobial effects of bleach are largely based on HOCls ability to cause aggregation of essential bacterial proteins.


PLOS Biology | 2004

Protein Thiol Modifications Visualized In Vivo

Lars I. Leichert; Ursula Jakob

Thiol-disulfide interconversions play a crucial role in the chemistry of biological systems. They participate in the major systems that control the cellular redox potential and prevent oxidative damage. In addition, thiol-disulfide exchange reactions serve as molecular switches in a growing number of redox-regulated proteins. We developed a differential thiol-trapping technique combined with two-dimensional gel analysis, which in combination with genetic studies, allowed us to obtain a snapshot of the in vivo thiol status of cellular proteins. We determined the redox potential of protein thiols in vivo, identified and dissected the in vivo substrate proteins of the major cellular thiol-disulfide oxidoreductases, and discovered proteins that undergo thiol modifications during oxidative stress. Under normal growth conditions most cytosolic proteins had reduced cysteines, confirming existing dogmas. Among the few partly oxidized cytosolic proteins that we detected were proteins that are known to form disulfide bond intermediates transiently during their catalytic cycle (e.g., dihydrolipoyl transacetylase and lipoamide dehydrogenase). Most proteins with highly oxidized thiols were periplasmic proteins and were found to be in vivo substrates of the disulfide-bond-forming protein DsbA. We discovered a substantial number of redox-sensitive cytoplasmic proteins, whose thiol groups were significantly oxidized in strains lacking thioredoxin A. These included detoxifying enzymes as well as many metabolic enzymes with active-site cysteines that were not known to be substrates for thioredoxin. H2O2-induced oxidative stress resulted in the specific oxidation of thiols of proteins involved in detoxification of H2O2 and of enzymes of cofactor and amino acid biosynthesis pathways such as thiolperoxidase, GTP-cyclohydrolase I, and the cobalamin-independent methionine synthase MetE. Remarkably, a number of these proteins were previously or are now shown to be redox regulated.


Molecular Cell | 2000

RNA Methylation under Heat Shock Control

Hans Bügl; Eric B. Fauman; Bart L. Staker; Fuzhong Zheng; Sidney R. Kushner; Mark A. Saper; James C. A. Bardwell; Ursula Jakob

Structural, biochemical, and genetic techniques were applied to investigate the function of FtsJ, a recently identified heat shock protein. FtsJ is well conserved, from bacteria to humans. The 1.5 A crystal structure of FtsJ in complex with its cofactor S-adenosylmethionine revealed that FtsJ has a methyltransferase fold. The molecular surface of FtsJ exposes a putative nucleic acid binding groove composed of highly conserved, positively charged residues. Substrate analysis showed that FtsJ methylates 23S rRNA within 50S ribosomal subunits in vitro and in vivo. Null mutations in ftsJ show a dramatically altered ribosome profile, a severe growth disadvantage, and a temperature-sensitive phenotype. Our results reveal an unexpected link between the heat shock response and RNA metabolism.


Methods in Enzymology | 1998

ANALYSIS OF CHAPERONE FUNCTION USING CITRATE SYNTHASE AS NONNATIVE SUBSTRATE PROTEIN

Johannes Buchner; Holger Grallert; Ursula Jakob

Publisher Summary The chapter presents a study related to the analysis of chaperone function by using citrate synthase (CS) as nonnative substrate protein. The chapter describes the use of CS as a chaperone substrate. CS is well suited, because unfolding and refolding can be easily monitored and both the thermal unfolding, as well as the folding pathway of chemically denatured CS is known in some detail. The influence of the four major classes of molecular chaperones—small heat shock proteins (Hsps), GroEL, Hsp70, and Hsp90—on CS folding has been characterized. The use of CS as a substrate protein for molecular chaperones allows one to address whether chaperones suppress aggregation, undergo stable or transient interactions with the substrate protein, interact with early- or late-unfolding intermediates, act in an adenosine triphosphate (ATP) -dependent manner, and resemble a member of one of the major classes of molecular chaperones. Citrate synthase (EC 4.1.3.7) is a commercially available, dimeric, mitochondrial protein, composed of two identical subunits (48.969 kDa each). The thermal unfolding of CS can be induced in vitro at temperatures similar to heat shock temperatures in vivo. Depending on the functional mechanism of the individual chaperone, aggregation can be affected differently.


Journal of Biological Chemistry | 2013

Oxidant Sensing by Reversible Disulfide Bond Formation

Claudia M. Cremers; Ursula Jakob

Maintenance of the cellular redox balance is crucial for cell survival. An increase in reactive oxygen, nitrogen, or chlorine species can lead to oxidative stress conditions, potentially damaging DNA, lipids, and proteins. Proteins are very sensitive to oxidative modifications, particularly methionine and cysteine residues. The reversibility of some of these oxidative protein modifications makes them ideally suited to take on regulatory roles in protein function. This is especially true for disulfide bond formation, which has the potential to mediate extensive yet fully reversible structural and functional changes, rapidly adjusting the proteins activity to the prevailing oxidant levels.


The EMBO Journal | 1994

Stress- and mitogen-induced phosphorylation of the small heat shock protein Hsp25 by MAPKAP kinase 2 is not essential for chaperone properties and cellular thermoresistance.

Knauf U; Ursula Jakob; K Engel; Johannes Buchner; Matthias Gaestel

Small heat shock proteins (sHsps) show a very rapid stress‐ and mitogen‐dependent phosphorylation by MAPKAP kinase 2. Based on this observation, phosphorylation of sHsps was thought to play a key role in mediating thermoresistance immediately after heat shock, before the increased synthesis of heat shock proteins becomes relevant. We have analysed the phosphorylation dependence of the chaperone and thermoresistance‐mediating properties of the small heat shock protein Hsp25. Surprisingly, overexpression of Hsp25 mutants, which are not phosphorylated in the transfected cells, confers the same thermoresistant phenotype as overexpression of wild type Hsp25, which is either mono‐ or bis‐phosphorylated at serine residues 15 and 86 within the cells. Furthermore, in vitro phosphorylated Hsp25 shows the same oligomerization properties and the same chaperone activity as the nonphosphorylated protein. No differences between phosphorylated and nonphosphorylated Hsp25 are detected in preventing thermal aggregation of unfolding proteins and assisting refolding of denatured proteins. The results suggest that chaperone properties of the small heat shock proteins contribute to the increased cellular thermoresistance in a phosphorylation‐independent manner.


Nature Structural & Molecular Biology | 2007

The redox-switch domain of Hsp33 functions as dual stress sensor

Marianne Ilbert; Janina Horst; Sebastian Ahrens; Jeannette Winter; Paul C.F. Graf; Hauke Lilie; Ursula Jakob

The redox-regulated chaperone Hsp33 is specifically activated upon exposure of cells to peroxide stress at elevated temperatures. Here we show that Hsp33 harbors two interdependent stress-sensing regions located in the C-terminal redox-switch domain of Hsp33: a zinc center sensing peroxide stress conditions and an adjacent linker region responding to unfolding conditions. Neither of these sensors works sufficiently in the absence of the other, making the simultaneous presence of both stress conditions a necessary requirement for Hsp33s full activation. Upon activation, Hsp33s redox-switch domain adopts a natively unfolded conformation, thereby exposing hydrophobic surfaces in its N-terminal substrate-binding domain. The specific activation of Hsp33 by the oxidative unfolding of its redox-switch domain makes this chaperone optimally suited to quickly respond to oxidative stress conditions that lead to protein unfolding.

Collaboration


Dive into the Ursula Jakob's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dana Reichmann

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge