92 results on '"Dorothee Kern"'
Search Results
2. Rescue of conformational dynamics in enzyme catalysis by directed evolution
- Author
-
Renee Otten, Lin Liu, Lillian R. Kenner, Michael W. Clarkson, David Mavor, Dan S. Tawfik, Dorothee Kern, and James S. Fraser
- Subjects
Science - Abstract
A key challenge in the field of protein design and evolution is to understand the mechanisms by which directed evolution is improving enzymes. Here the authors combine different biophysical methods and give mechanistic insights into how directed evolution increases the catalytic efficiency of human peptidyl-prolyl cis/trans isomerase CypA.
- Published
- 2018
- Full Text
- View/download PDF
3. Dynamics of human protein kinase Aurora A linked to drug selectivity
- Author
-
Warintra Pitsawong, Vanessa Buosi, Renee Otten, Roman V Agafonov, Adelajda Zorba, Nadja Kern, Steffen Kutter, Gunther Kern, Ricardo AP Pádua, Xavier Meniche, and Dorothee Kern
- Subjects
protein kinase ,drug binding ,enzyme kinetics ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Protein kinases are major drug targets, but the development of highly-selective inhibitors has been challenging due to the similarity of their active sites. The observation of distinct structural states of the fully-conserved Asp-Phe-Gly (DFG) loop has put the concept of conformational selection for the DFG-state at the center of kinase drug discovery. Recently, it was shown that Gleevec selectivity for the Tyr-kinase Abl was instead rooted in conformational changes after drug binding. Here, we investigate whether protein dynamics after binding is a more general paradigm for drug selectivity by characterizing the binding of several approved drugs to the Ser/Thr-kinase Aurora A. Using a combination of biophysical techniques, we propose a universal drug-binding mechanism, that rationalizes selectivity, affinity and long on-target residence time for kinase inhibitors. These new concepts, where protein dynamics in the drug-bound state plays the crucial role, can be applied to inhibitor design of targets outside the kinome.
- Published
- 2018
- Full Text
- View/download PDF
4. Conformational Selection in a Protein-Protein Interaction Revealed by Dynamic Pathway Analysis
- Author
-
Kalyan S. Chakrabarti, Roman V. Agafonov, Francesco Pontiggia, Renee Otten, Matthew K. Higgins, Gebhard F.X. Schertler, Daniel D. Oprian, and Dorothee Kern
- Subjects
conformational ensemble ,conformational selection ,energy landscape ,molecular recognition dynamics ,protein/protein interaction ,recoverin ,Biology (General) ,QH301-705.5 - Abstract
Molecular recognition plays a central role in biology, and protein dynamics has been acknowledged to be important in this process. However, it is highly debated whether conformational changes happen before ligand binding to produce a binding-competent state (conformational selection) or are caused in response to ligand binding (induced fit). Proposals for both mechanisms in protein/protein recognition have been primarily based on structural arguments. However, the distinction between them is a question of the probabilities of going via these two opposing pathways. Here, we present a direct demonstration of exclusive conformational selection in protein/protein recognition by measuring the flux for rhodopsin kinase binding to its regulator recoverin, an important molecular recognition in the vision system. Using nuclear magnetic resonance (NMR) spectroscopy, stopped-flow kinetics, and isothermal titration calorimetry, we show that recoverin populates a minor conformation in solution that exposes a hydrophobic binding pocket responsible for binding rhodopsin kinase. Protein dynamics in free recoverin limits the overall rate of binding.
- Published
- 2016
- Full Text
- View/download PDF
5. Molecular mechanism of Aurora A kinase autophosphorylation and its allosteric activation by TPX2
- Author
-
Adelajda Zorba, Vanessa Buosi, Steffen Kutter, Nadja Kern, Francesco Pontiggia, Young-Jin Cho, and Dorothee Kern
- Subjects
kinase ,activation ,mechanism ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
We elucidate the molecular mechanisms of two distinct activation strategies (autophosphorylation and TPX2-mediated activation) in human Aurora A kinase. Classic allosteric activation is in play where either activation loop phosphorylation or TPX2 binding to a conserved hydrophobic groove shifts the equilibrium far towards the active conformation. We resolve the controversy about the mechanism of autophosphorylation by demonstrating intermolecular autophosphorylation in a long-lived dimer by combining X-ray crystallography with functional assays. We then address the allosteric activation by TPX2 through activity assays and the crystal structure of a domain-swapped dimer of dephosphorylated Aurora A and TPX21−25. While autophosphorylation is the key regulatory mechanism in the centrosomes in the early stages of mitosis, allosteric activation by TPX2 of dephosphorylated Aurora A could be at play in the spindle microtubules. The mechanistic insights into autophosphorylation and allosteric activation by TPX2 binding proposed here, may have implications for understanding regulation of other protein kinases.
- Published
- 2014
- Full Text
- View/download PDF
6. Structure determination of high-energy states in a dynamic protein ensemble
- Author
-
John B. Stiller, Renee Otten, Daniel Häussinger, Pascal S. Rieder, Douglas L. Theobald, and Dorothee Kern
- Subjects
Multidisciplinary ,Protein Conformation ,Adenylate Kinase ,Cryoelectron Microscopy ,Thermodynamics ,Crystallography, X-Ray ,Nuclear Magnetic Resonance, Biomolecular ,Article - Abstract
Macromolecular function frequently requires that proteins change conformation into high-energy states(1–4). However, methods for solving the structures of these functionally essential, lowly populated states are lacking. Here we develop a method for high-resolution structure determination of minorly populated states by coupling NMR spectroscopy-derived pseudocontact shifts(5) (PCSs) with Carr–Purcell–Meiboom–Gill (CPMG) relaxation dispersion(6) (PCS–CPMG). Our approach additionally defines the corresponding kinetics and thermodynamics of high-energy excursions, thereby characterizing the entire free-energy landscape. Using a large set of simulated data for adenylate kinase (Adk), calmodulin and Src kinase, we find that high-energy PCSs accurately determine high-energy structures (with a root mean squared deviation of less than 3.5 angström). Applying our methodology to Adk during catalysis, we find that the high-energy excursion involves surprisingly small openings of the AMP and ATP lids. This previously unresolved high-energy structure solves a longstanding controversy about conformational interconversions that are rate-limiting for catalysis. Primed for either substrate binding or product release, the high-energy structure of Adk suggests a two-step mechanism combining conformational selection to this state, followed by an induced-fit step into a fully closed state for catalysis of the phosphoryl-transfer reaction. Unlike other methods for resolving high-energy states, such as cryo-electron microscopy and X-ray crystallography, our solution PCS–CPMG approach excels in cases involving domain rearrangements of smaller systems (less than 60 kDa) and populations as low as 0.5%, and enables the simultaneous determination of protein structure, kinetics and thermodynamics while proteins perform their function.
- Published
- 2022
- Full Text
- View/download PDF
7. From primordial clocks to circadian oscillators
- Author
-
Warintra Pitsawong, Ricardo A. P. Pádua, Timothy Grant, Marc Hoemberger, Renee Otten, Niels Bradshaw, Nikolaus Grigorieff, and Dorothee Kern
- Subjects
Multidisciplinary - Abstract
Circadian rhythms play an essential part in many biological processes, and only three prokaryotic proteins are required to constitute a true post-translational circadian oscillator1. The evolutionary history of the three Kai proteins indicates that KaiC is the oldest member and a central component of the clock2. Subsequent additions of KaiB and KaiA regulate the phosphorylation state of KaiC for time synchronization. The canonical KaiABC system in cyanobacteria is well understood3–6, but little is known about more ancient systems that only possess KaiBC. However, there are reports that they might exhibit a basic, hourglass-like timekeeping mechanism7–9. Here we investigate the primordial circadian clock in Rhodobacter sphaeroides, which contains only KaiBC, to elucidate its inner workings despite missing KaiA. Using a combination of X-ray crystallography and cryogenic electron microscopy, we find a new dodecameric fold for KaiC, in which two hexamers are held together by a coiled-coil bundle of 12 helices. This interaction is formed by the carboxy-terminal extension of KaiC and serves as an ancient regulatory moiety that is later superseded by KaiA. A coiled-coil register shift between daytime and night-time conformations is connected to phosphorylation sites through a long-range allosteric network that spans over 140 Å. Our kinetic data identify the difference in the ATP-to-ADP ratio between day and night as the environmental cue that drives the clock. They also unravel mechanistic details that shed light on the evolution of self-sustained oscillators.
- Published
- 2022
- Full Text
- View/download PDF
8. Prediction of multiple conformational states by combining sequence clustering with AlphaFold2
- Author
-
Hannah K. Wayment-Steele, Sergey Ovchinnikov, Lucy Colwell, and Dorothee Kern
- Abstract
AlphaFold2 (AF2) has revolutionized structural biology by accurately predicting single structures of proteins and protein-protein complexes. However, biological function is rooted in a protein’s ability to sample different conformational substates, and disease-causing point mutations are often due to population changes of these substates. This has sparked immense interest in expanding AF2’s capability to predict conformational substates. We demonstrate that clustering an input multiple sequence alignment (MSA) by sequence similarity enables AF2 to sample alternate states of known metamorphic proteins, including the circadian rhythm protein KaiB, the transcription factor RfaH, and the spindle checkpoint protein Mad2, and score these states with high confidence. Moreover, we use AF2 to identify a minimal set of two point mutations predicted to switch KaiB between its two states. Finally, we used our clustering method, AF-cluster, to screen for alternate states in protein families without known fold-switching, and identified a putative alternate state for the oxidoreductase DsbE. Similarly to KaiB, DsbE is predicted to switch between a thioredoxin-like fold and a novel fold. This prediction is the subject of future experimental testing. Further development of such bioinformatic methods in tandem with experiments will likely have profound impact on predicting protein energy landscapes, essential for shedding light into biological function.
- Published
- 2022
- Full Text
- View/download PDF
9. How directed evolution reshapes the energy landscape in an enzyme to boost catalysis
- Author
-
Aina E. Cohen, Vy Nguyen, Shuo Sui, Warintra Pitsawong, Sarah L. Perry, Renee Otten, Donald Hilvert, Dorothee Kern, H. Adrian Bunzel, Ricardo A.P. de Pádua, and MacKenzie Patterson
- Subjects
chemistry.chemical_classification ,Multidisciplinary ,Protein Conformation ,Extramural ,Catalytic function ,Proteins ,Energy landscape ,Directed evolution ,Enzymes ,Catalysis ,Enzyme ,Protein structure ,chemistry ,Chemical physics ,Catalytic Domain ,Biocatalysis ,Computer-Aided Design ,Directed Molecular Evolution ,Nuclear Magnetic Resonance, Biomolecular - Abstract
Two steps forward—now look back Whether designed computationally or uncovered in activity screening, enzymes repurposed for biocatalysis rarely start at the peak of proficiency. However, directed evolution can in some cases increase catalytic efficiency of a poor enzyme by many orders of magnitude. Otten et al. used a suite of biochemical techniques to investigate the origins of rate enhancement in a previously evolved model enzyme. Two conformational states are present in the initial, computationally designed enzyme, but only one is active. Shifting the population toward the active state is one factor in increasing catalytic efficiency during evolution. Single mutations do not greatly increase activity, but the synergistic combination of just two out of 17 substitutions can provide most of the rate enhancement seen in the final, evolved enzyme. Science , this issue p. 1442
- Published
- 2020
- Full Text
- View/download PDF
10. Cumulative mechanism of several major imatinib-resistant mutations in Abl kinase
- Author
-
Warintra Pitsawong, Dorothee Kern, and Marc Hoemberger
- Subjects
0301 basic medicine ,Antineoplastic Agents ,Drug resistance ,03 medical and health sciences ,chemistry.chemical_compound ,Adenosine Triphosphate ,0302 clinical medicine ,Neoplasms ,hemic and lymphatic diseases ,medicine ,Humans ,Amino Acid Sequence ,Kinase activity ,Oncogene Proteins v-abl ,Multidisciplinary ,ABL ,Kinase ,Imatinib ,Biological Sciences ,030104 developmental biology ,Protein kinase domain ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Imatinib Mesylate ,Cancer research ,Adenosine triphosphate ,Tyrosine kinase ,medicine.drug - Abstract
Despite the outstanding success of the cancer drug imatinib, one obstacle in prolonged treatment is the emergence of resistance mutations within the kinase domain of its target, Abl. We noticed that many patient-resistance mutations occur in the dynamic hot spots recently identified to be responsible for imatinib’s high selectivity toward Abl. In this study, we provide an experimental analysis of the mechanism underlying drug resistance for three major resistance mutations (G250E, Y253F, and F317L). Our data settle controversies, revealing unexpected resistance mechanisms. The mutations alter the energy landscape of Abl in complex ways: increased kinase activity, altered affinity, and cooperativity for the substrates, and, surprisingly, only a modestly decreased imatinib affinity. Only under cellular adenosine triphosphate (ATP) concentrations, these changes cumulate in an order of magnitude increase in imatinib’s half-maximal inhibitory concentration (IC(50)). These results highlight the importance of characterizing energy landscapes of targets and its changes by drug binding and by resistance mutations developed by patients.
- Published
- 2020
- Full Text
- View/download PDF
11. Ancient origins of allosteric activation in a Ser-Thr kinase
- Author
-
Warintra Pitsawong, Vy Nguyen, Adelajda Hadzipasic, Nadja Kern, Dorothee Kern, Janice Villali, Yuejiao Zheng, Christine D. Wilson, and Chansik Kim
- Subjects
Enzyme activator ,Multidisciplinary ,Kinase ,Targeting Protein for Xklp2 ,Autophosphorylation ,Allosteric regulation ,Aurora A kinase ,Colocalization ,Kinase binding ,Biology ,Cell biology - Abstract
Evolution of a kinase allosteric site Enzyme activity is often regulated by conformational changes coupled to binding of an effector at an allosteric site, a feature especially important for enzymes involved in signaling cascades. Hadzipasic et al. studied the origins of allosteric regulation of Aurora A, a kinase involved in progression of the eukaryotic cell cycle. Aurora A is allosterically regulated through the binding of an effector protein named TPX2, which also targets the kinase to spindle microtubules. By reconstructing ancestor kinase sequences, they found that TPX2 bound to an early Aurora A but had very weak activation that was gradually strengthened by evolution of an allosteric network within the kinase. An evolutionary advantage from localizing the active protein at the mitotic spindle may have driven the development of this regulatory mechanism. Science , this issue p. 912
- Published
- 2020
- Full Text
- View/download PDF
12. Probing the transition state in enzyme catalysis by high-pressure NMR dynamics
- Author
-
Marc Hoemberger, Dorothee Kern, Renee Otten, Young-Jin Cho, S. Jordan Kerns, Michael F. Hagan, and John B. Stiller
- Subjects
Quantitative Biology::Biomolecules ,Transition (genetics) ,Chemistry ,Quantitative Biology::Molecular Networks ,Process Chemistry and Technology ,digestive, oral, and skin physiology ,Kinetics ,Solvation ,Energy landscape ,Bioengineering ,Biochemistry ,Article ,Catalysis ,Transition state ,Enzyme catalysis ,Molecular dynamics ,Chemical physics - Abstract
Protein conformational changes are frequently essential for enzyme catalysis, and in several cases, shown to be the limiting factor for overall catalytic speed. However, a structural understanding of corresponding transition states, needed to rationalize the kinetics, remains obscure due to their fleeting nature. Here, we determine the transition-state ensemble of the rate-limiting conformational transition in the enzyme adenylate kinase, by a synergistic approach between experimental high-pressure NMR relaxation during catalysis and molecular dynamics simulations. By comparing homologous kinases evolved under ambient or high pressure in the deep-sea, we detail transition state ensembles that differ in solvation as directly measured by the pressure dependence of catalysis. Capturing transition-state ensembles begins to complete the catalytic energy landscape that is generally characterized by structures of all intermediates and frequencies of transitions among them.
- Published
- 2019
- Full Text
- View/download PDF
13. Bimodal evolution of Src and Abl kinase substrate specificity revealed using mammalian cell extract as substrate pool
- Author
-
Douglas L. Theobald, Danielle L. Swaney, Christine D. Wilson, Vanessa Buosi, Renee Otten, Margaret Soucheray, Dorothee Kern, Nevan J. Krogan, and Patrick Finneran
- Subjects
ABL ,Kinase ,Chemistry ,Biological signal transduction ,Phosphorylation ,Neofunctionalization ,Signal transduction ,Tyrosine kinase ,Cell biology ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The specificity of phosphorylation by protein kinases is essential to the integrity of biological signal transduction. While peptide sequence specificity for individual kinases has been examined previously, here we explore the evolutionary progression that has led to the modern substrate specificity of two non-receptor tyrosine kinases, Abl and Src. To efficiently determine the substrate specificity of modern and reconstructed ancestral kinases, we developed a method using mammalian cell lysate as the substrate pool, thereby representing the naturally occurring substrate proteins. We find that the oldest tyrosine kinase ancestor was a promiscuous enzyme that evolved through a more specific last common ancestor into a specific human Abl. In contrast, the parallel pathway to human Src involved a loss of substrate specificity, leading to general promiscuity. These results add a new facet to our understanding of the evolution of signaling pathways, with both subfunctionalization and neofunctionalization along the evolutionary trajectories.
- Published
- 2020
- Full Text
- View/download PDF
14. Decision letter: Breakage of the oligomeric CaMKII hub by the regulatory segment of the kinase
- Author
-
Richard Bayliss and Dorothee Kern
- Subjects
Breakage ,Kinase ,Chemistry ,Ca2+/calmodulin-dependent protein kinase ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
15. From structure to mechanism: skiing the energy landscape
- Author
-
Dorothee Kern
- Subjects
Structure (mathematical logic) ,0303 health sciences ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Protein Conformation ,Computer science ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Environmental resource management ,Proteins ,Energy landscape ,Cell Biology ,Biochemistry ,ComputingMilieux_GENERAL ,03 medical and health sciences ,ComputingMethodologies_PATTERNRECOGNITION ,Thermodynamics ,business ,Molecular Biology ,Mechanism (sociology) ,030304 developmental biology ,Biotechnology - Abstract
It is time for structural biologists to embrace the challenge of quantitatively describing functional energy landscapes.
- Published
- 2021
- Full Text
- View/download PDF
16. Allosteric modulation of a human protein kinase with monobodies
- Author
-
Adelajda Zorba, Shohei Koide, Marc Hoemberger, Akiko Koide, Chansik Kim, Vy Nguyen, Dorothee Kern, Yuejiao Zheng, and Steffen Kutter
- Subjects
0301 basic medicine ,Protein Conformation ,Fibronectin Type III Domain ,Allosteric regulation ,Drug design ,Crystallography, X-Ray ,03 medical and health sciences ,0302 clinical medicine ,Adenosine Triphosphate ,Allosteric Regulation ,Aurora Kinase B ,Humans ,Kinase activity ,Binding site ,Protein kinase A ,Structural motif ,Protein Kinase Inhibitors ,Aurora Kinase A ,Multidisciplinary ,Binding Sites ,Kinase ,Chemistry ,Biological Sciences ,Monobody ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Drug Design ,Carrier Proteins ,Protein Kinases - Abstract
Despite being the subject of intense effort and scrutiny, kinases have proven to be consistently challenging targets in inhibitor drug design. A key obstacle has been promiscuity and consequent adverse effects of drugs targeting the ATP binding site. Here we introduce an approach to controlling kinase activity by using monobodies that bind to the highly specific regulatory allosteric pocket of the oncoprotein Aurora A (AurA) kinase, thereby offering the potential for more specific kinase modulators. Strikingly, we identify a series of highly specific monobodies acting either as strong kinase inhibitors or activators via differential recognition of structural motifs in the allosteric pocket. X-ray crystal structures comparing AurA bound to activating vs inhibiting monobodies reveal the atomistic mechanism underlying allosteric modulation. The results reveal 3 major advantages of targeting allosteric vs orthosteric sites: extreme selectivity, ability to inhibit as well as activate, and avoidance of competing with ATP that is present at high concentrations in the cells. We envision that exploiting allosteric networks for inhibition or activation will provide a general, powerful pathway toward rational drug design.
- Published
- 2019
17. Evolutionary drivers of thermoadaptation in enzyme catalysis
- Author
-
Vy Nguyen, Justin English, Douglas L. Theobald, Marc Hoemberger, Dorothee Kern, John B. Stiller, Roman V. Agafonov, Steffen Kutter, and Christine D. Wilson
- Subjects
Thermotolerance ,0301 basic medicine ,Hot Temperature ,Multidisciplinary ,Sequence reconstruction ,biology ,Adenylate kinase ,Article ,Early life ,Enzyme assay ,Enzyme catalysis ,Evolution, Molecular ,Kinetics ,03 medical and health sciences ,030104 developmental biology ,Molecular level ,Biochemistry ,Phylogenetics ,Evolutionary biology ,Mutation ,Biocatalysis ,biology.protein ,Psychrophile ,Phylogeny ,Adenylyl Cyclases - Abstract
With early life likely to have existed in a hot environment, enzymes had to cope with an inherent drop in catalytic speed caused by lowered temperature. Here we characterize the molecular mechanisms underlying thermoadaptation of enzyme catalysis in adenylate kinase using ancestral sequence reconstruction spanning 3 billion years of evolution. We show that evolution solved the enzyme’s key kinetic obstacle—how to maintain catalytic speed on a cooler Earth—by exploiting transition-state heat capacity. Tracing the evolution of enzyme activity and stability from the hot-start toward modern hyperthermophilic, mesophilic, and psychrophilic organisms illustrates active pressure versus passive drift in evolution on a molecular level, refutes the debated activity/stability trade-off, and suggests that the catalytic speed of adenylate kinase is an evolutionary driver for organismal fitness.
- Published
- 2017
- Full Text
- View/download PDF
18. Regulation of Microtubule Assembly by Tau and not by Pin1
- Author
-
Alexandra M. Deaconescu, Dorothee Kern, Steffen Kutter, and Timo Eichner
- Subjects
0301 basic medicine ,Magnetic Resonance Spectroscopy ,tau Proteins ,macromolecular substances ,Isomerase ,Microtubules ,Epitope ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,Microtubule ,Scattering, Small Angle ,Humans ,Molecular Biology ,biology ,Chemistry ,Negative stain ,NIMA-Interacting Peptidylprolyl Isomerase ,Microscopy, Electron ,030104 developmental biology ,Förster resonance energy transfer ,Tubulin ,Biochemistry ,PIN1 ,biology.protein ,Biophysics ,Phosphorylation ,Protein Multimerization ,030217 neurology & neurosurgery - Abstract
The molecular mechanism by which the microtubule-associated protein (MAP) tau regulates the formation of microtubules (MTs) is poorly understood. The activity of tau is controlled via phosphorylation at specific Ser/Thr sites. Of those phosphorylation sites, 17 precede a proline, making them potential recognition sites for the peptidyl-prolyl isomerase Pin1. Pin1 binding and catalysis of phosphorylated tau at the AT180 epitope, which was implicated in Alzheimer's disease, has been reported to be crucial for restoring tau's ability to promote MT polymerization in vitro and in vivo [1]. Surprisingly, we discover that Pin1 does not promote phosphorylated tau-induced MT formation in vitro, refuting the commonly accepted model in which Pin1 binding and catalysis on the A180 epitope restores the function of the Alzheimer's associated phosphorylated tau in tubulin assembly [1, 2]. Using turbidity assays, time-resolved small angle X-ray scattering (SAXS), and time-resolved negative stain electron microscopy (EM), we investigate the mechanism of tau-mediated MT assembly and the role of the Thr231 and Ser235 phosphorylation on this process. We discover novel GTP-tubulin ring-shaped species, which are detectable in the earliest stage of tau-induced polymerization and may play a crucial role in the early nucleation phase of MT assembly. Finally, by NMR and SAXS experiments, we show that the tau molecules must be located on the surface of MTs and tubulin rings during the polymerization reaction. The interaction between tau and tubulin is multipartite, with a high affinity interaction of the four tubulin-binding repeats, and a weaker interaction with the proline-rich sequence and the termini of tau.
- Published
- 2016
- Full Text
- View/download PDF
19. Mechanism of activating mutations and allosteric drug inhibition of the phosphatase SHP2
- Author
-
Warintra Pitsawong, Yizhi Sun, Renee Otten, Ricardo A.P. de Pádua, Ingrid Marko, John B. Stiller, and Dorothee Kern
- Subjects
0301 basic medicine ,Magnetic Resonance Spectroscopy ,Protein Conformation ,Science ,Allosteric regulation ,Phosphatase ,Mutant ,Regulator ,General Physics and Astronomy ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,Protein tyrosine phosphatase ,Crystallography, X-Ray ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Protein structure ,Allosteric Regulation ,Piperidines ,Hydrolase ,medicine ,Humans ,lcsh:Science ,Mutation ,Multidisciplinary ,Chemistry ,General Chemistry ,Pyrimidines ,030104 developmental biology ,Biophysics ,lcsh:Q - Abstract
Protein tyrosine phosphatase SHP2 functions as a key regulator of cell cycle control, and activating mutations cause several cancers. Here, we dissect the energy landscape of wild-type SHP2 and the oncogenic mutation E76K. NMR spectroscopy and X-ray crystallography reveal that wild-type SHP2 exchanges between closed, inactive and open, active conformations. E76K mutation shifts this equilibrium toward the open state. The previously unknown open conformation is characterized, including the active-site WPD loop in the inward and outward conformations. Binding of the allosteric inhibitor SHP099 to E76K mutant, despite much weaker, results in an identical structure as the wild-type complex. A conformational selection to the closed state reduces drug affinity which, combined with E76K’s much higher activity, demands significantly greater SHP099 concentrations to restore wild-type activity levels. The differences in structural ensembles and drug-binding kinetics of cancer-associated SHP2 forms may stimulate innovative ideas for developing more potent inhibitors for activated SHP2 mutants., The protein tyrosine phosphatase SHP2 is a key regulator of cell cycle control. Here the authors combine NMR measurements and X-ray crystallography and show that wild-type SHP2 dynamically exchanges between a closed inactive conformation and an open activated form and that the oncogenic E76K mutation shifts the equilibrium to the open state, which is reversed by binding of the allosteric inhibitor SHP099.
- Published
- 2018
- Full Text
- View/download PDF
20. Decision letter: Ancestral reconstruction reveals mechanisms of ERK regulatory evolution
- Author
-
Dorothee Kern
- Subjects
MAPK/ERK pathway ,Ancestral reconstruction ,Evolutionary biology ,Biology - Published
- 2018
- Full Text
- View/download PDF
21. Author response: Dynamics of human protein kinase Aurora A linked to drug selectivity
- Author
-
Roman V. Agafonov, Xavier Meniche, Adelajda Zorba, Nadja Kern, Warintra Pitsawong, Ricardo Ap Pádua, Renee Otten, Gunther Kern, Vanessa Buosi, Dorothee Kern, and Steffen Kutter
- Subjects
Drug ,Chemistry ,media_common.quotation_subject ,Dynamics (mechanics) ,Protein kinase A ,Selectivity ,media_common ,Cell biology - Published
- 2018
- Full Text
- View/download PDF
22. Dynamics of human protein kinase Aurora A linked to drug selectivity
- Author
-
Roman V. Agafonov, Dorothee Kern, Xavier Meniche, Vanessa Buosi, Steffen Kutter, Gunther Kern, Warintra Pitsawong, Renee Otten, Adelajda Zorba, Nadja Kern, and Ricardo Ap Pádua
- Subjects
0301 basic medicine ,QH301-705.5 ,Protein Conformation ,Science ,Structural Biology and Molecular Biophysics ,Plasma protein binding ,Computational biology ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,01 natural sciences ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Protein structure ,drug binding ,enzyme kinetics ,Drug Discovery ,Humans ,Kinome ,Biology (General) ,Protein kinase A ,Protein Kinase Inhibitors ,Cancer Biology ,Aurora Kinase A ,General Immunology and Microbiology ,010405 organic chemistry ,Drug discovery ,Chemistry ,General Neuroscience ,E. coli ,protein kinase ,General Medicine ,0104 chemical sciences ,3. Good health ,Kinetics ,030104 developmental biology ,Imatinib mesylate ,Structural biology ,Imatinib Mesylate ,Medicine ,Research Article ,Protein Binding - Abstract
Protein kinases are major drug targets, but the development of highly-selective inhibitors has been challenging due to the similarity of their active sites. The observation of distinct structural states of the fully-conserved Asp-Phe-Gly (DFG) loop has put the concept of conformational selection for the DFG-state at the center of kinase drug discovery. Recently, it was shown that Gleevec selectivity for the Tyr-kinase Abl was instead rooted in conformational changes after drug binding. Here, we investigate whether protein dynamics after binding is a more general paradigm for drug selectivity by characterizing the binding of several approved drugs to the Ser/Thr-kinase Aurora A. Using a combination of biophysical techniques, we propose a universal drug-binding mechanism, that rationalizes selectivity, affinity and long on-target residence time for kinase inhibitors. These new concepts, where protein dynamics in the drug-bound state plays the crucial role, can be applied to inhibitor design of targets outside the kinome., eLife digest Protein kinases are a family of enzymes found in all living organisms. These enzymes help to control many biological processes, including cell division. When particular protein kinases do not work correctly, cells may start to divide uncontrollably, which can lead to cancer. One example is the kinase Aurora A, which is over-active in many common human cancers. As a result, researchers are currently trying to design drugs that reduce the activity of Aurora A in the hope that these could form new anticancer treatments. In general, drugs are designed to be as specific in their action as possible to reduce the risk of harmful side effects to the patient. Designing a drug that affects a single protein kinase, however, is difficult because there are hundreds of different kinases in the body, all with similar structures. Because drugs often work by binding to specific structural features, a drug that targets one protein kinase can often alter the activity of a large number of others too. Gleevec is a successful anti-leukemia drug that specifically works on one target kinase, producing minimal side effects. It was recently discovered that the drug works through a phenomenon called ‘induced fit’. This means that after the drug binds it causes a change in the enzyme’s overall shape that alters the activity of the enzyme. The shape change is complex, and so even small structural differences can change the effect of a particular drug. Do other drugs that target other protein kinases also produce induced fit effects? To find out, Pitsawong, Buosi, Otten, Agafonov et al. studied how three anti-cancer drugs interact with Aurora A: two drugs specifically designed to switch off Aurora A, and Gleevec (which does not target Aurora A). The two drugs that specifically target Aurora A were thought to work by targeting one structural feature of the enzyme. However, the biochemical and biophysical experiments performed by Pitsawong et al. revealed that these drugs instead work through an induced fit effect. By contrast, Gleevec did not trigger an induced fit on Aurora A and so bound less tightly to it. In light of these results, Pitsawong et al. suggest that future efforts to design drugs that target protein kinases should focus on exploiting the induced fit process. This will require more research into the structure of particular kinases.
- Published
- 2018
23. Rescue of conformational dynamics in enzyme catalysis by directed evolution
- Author
-
Lin Liu, Michael W. Clarkson, James S. Fraser, Lillian R. Kenner, Renee Otten, Dorothee Kern, David Mavor, and Dan S. Tawfik
- Subjects
0301 basic medicine ,Models, Molecular ,Magnetic Resonance Spectroscopy ,Proline ,Science ,Allosteric regulation ,General Physics and Astronomy ,Isomerase ,Biology ,010402 general chemistry ,Crystallography, X-Ray ,01 natural sciences ,General Biochemistry, Genetics and Molecular Biology ,Article ,Catalysis ,Enzyme catalysis ,Substrate Specificity ,Cyclophilin A ,03 medical and health sciences ,Models ,Catalytic Domain ,Escherichia coli ,Humans ,lcsh:Science ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Crystallography ,Chemistry ,Protein dynamics ,Rational design ,Temperature ,Energy landscape ,Molecular ,General Chemistry ,Directed evolution ,0104 chemical sciences ,030104 developmental biology ,Biochemistry ,Mutation ,Biophysics ,X-Ray ,lcsh:Q ,Directed Molecular Evolution ,Monte Carlo Method ,Function (biology) - Abstract
Rational design and directed evolution have proved to be successful approaches to increase catalytic efficiencies of both natural and artificial enzymes. Protein dynamics is recognized as important, but due to the inherent flexibility of biological macromolecules it is often difficult to distinguish which conformational changes are directly related to function. Here, we use directed evolution on an impaired mutant of the proline isomerase CypA and identify two second-shell mutations that partially restore its catalytic activity. We show both kinetically, using NMR spectroscopy, and structurally, by room-temperature X-ray crystallography, how local perturbations propagate through a large allosteric network to facilitate conformational dynamics. The increased catalysis selected for in the evolutionary screen is correlated with an accelerated interconversion between the two catalytically essential conformational sub-states, which are both captured in the high-resolution X-ray ensembles. Our data provide a glimpse of an evolutionary trajectory and show how subtle changes can fine-tune enzyme function., A key challenge in the field of protein design and evolution is to understand the mechanisms by which directed evolution is improving enzymes. Here the authors combine different biophysical methods and give mechanistic insights into how directed evolution increases the catalytic efficiency of human peptidyl-prolyl cis/trans isomerase CypA.
- Published
- 2018
- Full Text
- View/download PDF
24. Dynamics of human protein kinases linked to drug selectivity
- Author
-
Warintra Pitsawong, Vanessa Buosi, Renee Otten, Roman V. Agafonov, Adelajda Zorba, Nadja Kern, Steffen Kutter, Gunther Kern, Ricardo A. P. Pádua, Xavier Meniche, and Dorothee Kern
- Subjects
Drug ,Chemistry ,Kinase ,media_common.quotation_subject ,Biophysics ,Preprint ,Selectivity ,media_common - Abstract
Protein kinases are major drug targets, but the development of highly-selective inhibitors has been challenging due to the similarity of their active sites. The observation of distinct structural states of the fully-conserved Asp-Phe-Gly (DFG) loop has put the concept of conformational selection for the DFG-state at the center of kinase drug discovery. Recently, it was shown that Gleevec selectivity for the Tyr-kinases Abl was instead rooted in conformational changes after drug binding. Here, we investigate whether protein dynamics after binding is a more general paradigm for drug selectivity by characterizing the binding of several approved drugs to the Ser/Thr-kinase Aurora A. Using a combination of biophysical techniques, we propose a universal drug-binding mechanism, that rationalizes selectivity, affinity and long on-target residence time for kinase inhibitors. These new concepts, where protein dynamics in the drug-bound state plays the crucial role, can be applied to inhibitor design of targets outside the kinome.eLife digestThe Ser/Thr kinase Aurora A is an important target for the development of new anticancer therapies. A longstanding question is how to specifically and effectively inhibit only this kinase in a background of over 550 protein kinases with very similar structures. To this end, understanding the inhibition mechanism of Aurora A by different drugs is essential. Here, we characterize the kinetic mechanism of three distinct kinase drugs, Gleevec (Imatinib), Danusertib (PHA739358) and AT9283 (Pyrazol-4-yl Urea) for Aurora A. We show that inhibitor affinities do not rely exclusively on the recognition of a specific conformation of the Asp-Phe-Gly loop of the kinase. Our quantitative kinetics data put forward an opposing mechanism in which a slow conformational change after drug binding (i.e., induced-fit step) dictates drug affinity.
- Published
- 2018
- Full Text
- View/download PDF
25. Using ancient protein kinases to unravel a modern cancer drug's mechanism
- Author
-
Douglas L. Theobald, Dorothee Kern, D. Waterman, Christine D. Wilson, Jackson C. Halpin, Vanessa Buosi, Steffen Kutter, Marc Hoemberger, Roman V. Agafonov, Renee Otten, and Adelajda Zorba
- Subjects
Genetics ,Multidisciplinary ,Protein structure ,Imatinib mesylate ,Common descent ,Oncogene Proteins v-abl ,Phylogenetics ,Energy landscape ,Plasma protein binding ,Computational biology ,Biology ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Macromolecular function is rooted in energy landscapes, where sequence determines not a single structure but an ensemble of conformations. Hence, evolution modifies a protein’s function by altering its energy landscape. Here, we recreate the evolutionary pathway between two modern human oncogenes, Src and Abl, by reconstructing their common ancestors. Our evolutionary reconstruction combined with x-ray structures of the common ancestor and pre–steady-state kinetics reveals a detailed atomistic mechanism for selectivity of the successful cancer drug Gleevec. Gleevec affinity is gained during the evolutionary trajectory toward Abl and lost toward Src, primarily by shifting an induced-fit equilibrium that is also disrupted in the clinical T315I resistance mutation. This work reveals the mechanism of Gleevec specificity while offering insights into how energy landscapes evolve.
- Published
- 2015
- Full Text
- View/download PDF
26. The energy landscape of adenylate kinase during catalysis
- Author
-
Lien A. Phung, Dimitar V. Pachov, Dorothee Kern, Tom Alber, Renee Otten, Roman V. Agafonov, Francesco Pontiggia, Vu Hong Thai, Padraig Niall Murphy, Steffen Kutter, Young-Jin Cho, Michael F. Hagan, and S. Jordan Kerns
- Subjects
Models, Molecular ,Binding Sites ,Magnetic Resonance Spectroscopy ,biology ,Chemistry ,Kinase ,Adenylate Kinase ,Active site ,Energy landscape ,Adenylate kinase ,Crystallography, X-Ray ,Catalysis ,Cofactor ,Biochemistry ,Catalytic cycle ,Structural Biology ,Catalytic Domain ,biology.protein ,Biophysics ,Transferase ,Binding site ,Molecular Biology - Abstract
Kinases perform phosphoryl-transfer reactions in milliseconds; without enzymes, these reactions would take about 8,000 years under physiological conditions. Despite extensive studies, a comprehensive understanding of kinase energy landscapes, including both chemical and conformational steps, is lacking. Here we scrutinize the microscopic steps in the catalytic cycle of adenylate kinase, through a combination of NMR measurements during catalysis, pre-steady-state kinetics, molecular-dynamics simulations and crystallography of active complexes. We find that the Mg(2+) cofactor activates two distinct molecular events: phosphoryl transfer (>10(5)-fold) and lid opening (10(3)-fold). In contrast, mutation of an essential active site arginine decelerates phosphoryl transfer 10(3)-fold without substantially affecting lid opening. Our results highlight the importance of the entire energy landscape in catalysis and suggest that adenylate kinases have evolved to activate key processes simultaneously by precise placement of a single, charged and very abundant cofactor in a preorganized active site.
- Published
- 2015
- Full Text
- View/download PDF
27. A minor conformation of a lanthanide tag on adenylate kinase characterized by paramagnetic relaxation dispersion NMR spectroscopy
- Author
-
Wei-Min Liu, Roman V. Agafonov, Lien A. Phung, Renee Otten, Marcellus Ubbink, Mathias A. S. Hass, Jesika Schilder, and Dorothee Kern
- Subjects
Models, Molecular ,Lanthanide ,Protein Conformation ,Chemistry ,Protein dynamics ,Adenylate Kinase ,Relaxation (NMR) ,Nuclear magnetic resonance spectroscopy ,Lanthanoid Series Elements ,Biochemistry ,Paramagnetism ,Nuclear magnetic resonance ,Protein structure ,Chemical physics ,Dispersion (optics) ,Nuclear Magnetic Resonance, Biomolecular ,Spectroscopy ,Macromolecule - Abstract
NMR relaxation dispersion techniques provide a powerful method to study protein dynamics by characterizing lowly populated conformations that are in dynamic exchange with the major state. Paramagnetic NMR is a versatile tool for investigating the structures and dynamics of proteins. These two techniques were combined here to measure accurate and precise pseudocontact shifts of a lowly populated conformation. This method delivers valuable long-range structural restraints for higher energy conformations of macromolecules in solution. Another advantage of combining pseudocontact shifts with relaxation dispersion is the increase in the amplitude of dispersion profiles. Lowly populated states are often involved in functional processes, such as enzyme catalysis, signaling, and protein/protein interactions. The presented results also unveil a critical problem with the lanthanide tag used to generate paramagnetic relaxation dispersion effects in proteins, namely that the motions of the tag can interfere severely with the observation of protein dynamics. The two-point attached CLaNP-5 lanthanide tag was linked to adenylate kinase. From the paramagnetic relaxation dispersion only motion of the tag is observed. The data can be described accurately by a two-state model in which the protein-attached tag undergoes a 23° tilting motion on a timescale of milliseconds. The work demonstrates the large potential of paramagnetic relaxation dispersion and the challenge to improve current tags to minimize relaxation dispersion from tag movements.
- Published
- 2015
- Full Text
- View/download PDF
28. Catalysis, dynamics and stability of enzymes under extreme conditions
- Author
-
Dorothee Kern
- Subjects
chemistry.chemical_classification ,Enzyme ,Chemistry ,Chemical physics ,Dynamics (mechanics) ,Stability (probability) ,Catalysis - Published
- 2017
- Full Text
- View/download PDF
29. Energetic dissection of Gleevec's selectivity toward human tyrosine kinases
- Author
-
Vanessa Buosi, Renee Otten, Dorothee Kern, Christine D. Wilson, and Roman V. Agafonov
- Subjects
Antineoplastic Agents ,Biology ,Piperazines ,Article ,SH3 domain ,Receptor tyrosine kinase ,Cell Line ,Structural Biology ,Neoplasms ,Humans ,Proto-Oncogene Proteins c-abl ,Protein Kinase Inhibitors ,Molecular Biology ,ABL ,Kinase ,Protein Structure, Tertiary ,Kinetics ,Pyrimidines ,src-Family Kinases ,Imatinib mesylate ,Biochemistry ,Benzamides ,Imatinib Mesylate ,biology.protein ,Thermodynamics ,Tyrosine kinase ,Protein Binding ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Protein kinases are obvious drug targets against cancer, owing to their central role in cellular regulation. Since the discovery of Gleevec, a potent and specific inhibitor of Abl kinase, as a highly successful cancer therapeutic, the ability of this drug to distinguish between Abl and other tyrosine kinases such as Src has been intensely investigated but without much success. Using NMR and fast kinetics, we establish a new model that solves this longstanding question of how the two tyrosine kinases adopt almost identical structures when bound to Gleevec but have vastly different affinities. We show that, in contrast to all other proposed models, the origin of Abl's high affinity lies predominantly in a conformational change after binding. An energy landscape providing tight affinity via an induced fit and binding plasticity via a conformational-selection mechanism is likely to be general for many inhibitors.
- Published
- 2014
- Full Text
- View/download PDF
30. Antiparallel EmrE exports drugs by exchanging between asymmetric structures
- Author
-
Emma A. Morrison, Reza Vafabakhsh, Supratik Dutta, Taekjip Ha, Katherine A. Henzler-Wildman, Dorothee Kern, Arjun Bahl, Gregory T. DeKoster, and Michael W. Clarkson
- Subjects
Models, Molecular ,Protein Conformation ,Dimer ,Model lipid bilayer ,010402 general chemistry ,Antiparallel (biochemistry) ,01 natural sciences ,7. Clean energy ,Article ,Antiporters ,03 medical and health sciences ,chemistry.chemical_compound ,Protein structure ,Catalytic Domain ,Escherichia coli ,Fluorescence Resonance Energy Transfer ,Nuclear Magnetic Resonance, Biomolecular ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,Escherichia coli Proteins ,Active site ,Water ,Biological Transport ,0104 chemical sciences ,3. Good health ,Crystallography ,Förster resonance energy transfer ,Structural biology ,Pharmaceutical Preparations ,biology.protein ,Protein Multimerization - Abstract
Small multidrug resistance transporters provide an ideal system to study the minimal requirements for active transport. EmrE is one such transporter in Escherichia coli. It exports a broad class of polyaromatic cation substrates, thus conferring resistance to drug compounds matching this chemical description. However, a great deal of controversy has surrounded the topology of the EmrE homodimer. Here we show that asymmetric antiparallel EmrE exchanges between inward- and outward-facing states that are identical except that they have opposite orientation in the membrane. We quantitatively measure the global conformational exchange between these two states for substrate-bound EmrE in bicelles using solution NMR dynamics experiments. Forster resonance energy transfer reveals that the monomers within each dimer are antiparallel, and paramagnetic relaxation enhancement NMR experiments demonstrate differential water accessibility of the two monomers within each dimer. Our experiments reveal a 'dynamic symmetry' that reconciles the asymmetric EmrE structure with the functional symmetry of residues in the active site.
- Published
- 2011
31. Dissecting the Microscopic Steps of the Cyclophilin A Enzymatic Cycle on the Biological HIV-1 Capsid Substrate by NMR
- Author
-
Oscar Millet, Elan Z. Eisenmesser, Dorothee Kern, Magnus Wolf-Watz, Daryl A. Bosco, Michael W. Clarkson, and Wladimir Labeikovsky
- Subjects
Models, Molecular ,Protein Conformation ,Cypa ,Isomerase ,In Vitro Techniques ,Biology ,Substrate Specificity ,Cyclophilin A ,Structural Biology ,Catalytic Domain ,Prolyl isomerase ,Humans ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,chemistry.chemical_classification ,Mutagenesis ,biology.organism_classification ,Recombinant Proteins ,Kinetics ,Enzyme ,Biochemistry ,chemistry ,Capsid ,HIV-1 ,Capsid Proteins ,Mutant Proteins ,human activities ,Heteronuclear single quantum coherence spectroscopy - Abstract
Peptidyl-prolyl isomerases (PPIases) are emerging as key regulators of many diverse biological processes. Elucidating the role of PPIase activity in vivo has been challenging because mutagenesis of active-site residues not only reduces the catalytic activity of these enzymes but also dramatically affects substrate binding. Employing the cyclophilin A PPIase together with its biologically relevant and natively folded substrate, the N-terminal domain of the human immunodeficiency virus type 1 capsid (CA(N)) protein, we demonstrate here how to dissect residue-specific contributions to PPIase catalysis versus substrate binding utilizing NMR spectroscopy. Surprisingly, a number of cyclophilin A active-site mutants previously assumed to be strongly diminished in activity toward biological substrates based only on a peptide assay catalyze the human immunodeficiency virus capsid with wild-type activity but with a change in the rate-limiting step of the enzymatic cycle. The results illustrate that a quantitative analysis of catalysis using the biological substrates is critical when interpreting the effects of PPIase mutations in biological assays.
- Published
- 2010
- Full Text
- View/download PDF
32. Choreographing an enzyme's dance
- Author
-
Janice Villali and Dorothee Kern
- Subjects
Component (thermodynamics) ,Stereochemistry ,Chemistry ,Energy landscape ,Kinetic energy ,Biochemistry ,Article ,Enzymes ,Analytical Chemistry ,Enzyme catalysis ,Catalysis ,Kinetics ,Purine-Nucleoside Phosphorylase ,Chemical physics ,Kinetic isotope effect ,Biocatalysis ,Animals ,Humans ,Thermodynamics ,Enzyme kinetics ,Ground state ,Hydrogen - Abstract
While ground state structures combined with chemical tools and enzyme kinetics deliver useful information on possible chemical mechanisms of enzyme catalysis, they do not unravel the finely balanced energy inventory to explain the impressive rate enhancement of enzymes. For this goal, a complete description of enzyme catalysis in the form of an energy landscape is needed. Since the rate of catalysis is determined by the climb over a sequence of energy barriers, we focus here on the critical question of transition pathways. A combination of time-resolved NMR and simulation deliver a glimpse into how proteins can so efficiently move within the ensemble of the native conformations while avoiding unfolding during that journey. The loss of energy due to breakage of native contacts is compensated by non-native transient hydrogen bonds during the transition thereby 'holding on' to the energy until the new native contacts form that define the alternate functional state. The use of kinetic isotope effects (KIE) to study the chemical step show that coordinated atomic fluctuations of the protein component dictate the probability of 'correct' distance and orientation, due to its extreme sensitivity to distance. The examples here stress the point that highly choreographed conformational sampling together with chemical integrity is a prerequisite for efficient enzyme catalysis.
- Published
- 2010
- Full Text
- View/download PDF
33. Peptidyl-Prolyl Isomerase FKBP52 Controls Chemotropic Guidance of Neuronal Growth Cones via Regulation of TRPC1 Channel Opening
- Author
-
Shmuel Muallem, Aleksandr Milshteyn, Paul F. Worley, Sangwoo Shim, Dorothee Kern, Guo Li Ming, Weizhong Zeng, Guo N. Huang, Ju Young Kim, and Joseph P. Yuan
- Subjects
Chemoautotrophic Growth ,Xenopus ,Neuroscience(all) ,Growth Cones ,Molecular Sequence Data ,DEVBIO ,Gating ,Biology ,Article ,MOLNEURO ,Cell Line ,Tacrolimus Binding Proteins ,03 medical and health sciences ,0302 clinical medicine ,Immunophilins ,Cell Movement ,Prolyl isomerase ,Animals ,Humans ,Amino Acid Sequence ,HSP90 Heat-Shock Proteins ,Growth cone ,Cells, Cultured ,TRPC Cation Channels ,030304 developmental biology ,Neurons ,Peptidylprolyl isomerase ,0303 health sciences ,General Neuroscience ,Peptidylprolyl Isomerase ,FKBP52 ,Rats ,3. Good health ,Cell biology ,FKBP ,Biochemistry ,Axon guidance ,CELLBIO ,Ion Channel Gating ,030217 neurology & neurosurgery - Abstract
SummaryImmunophilins, including FK506-binding proteins (FKBPs), are protein chaperones with peptidyl-prolyl isomerase (PPIase) activity. Initially identified as pharmacological receptors for immunosuppressants to regulate immune responses via isomerase-independent mechanisms, FKBPs are most highly expressed in the nervous system, where their physiological function as isomerases remains unknown. We demonstrate that FKBP12 and FKBP52 catalyze cis/trans isomerization of regions of TRPC1 implicated in controlling channel opening. FKBP52 mediates stimulus-dependent TRPC1 gating through isomerization, which is required for chemotropic turning of neuronal growth cones to netrin-1 and myelin-associated glycoprotein and for netrin-1/DCC-dependent midline axon guidance of commissural interneurons in the developing spinal cord. By contrast, FKBP12 mediates spontaneous opening of TRPC1 through isomerization and is not required for growth cone responses to netrin-1. Our study demonstrates a novel physiological function of proline isomerases in chemotropic nerve guidance through TRPC1 gating and may have significant implication in clinical applications of immunophilin-related therapeutic drugs.
- Published
- 2009
- Full Text
- View/download PDF
34. Segmented Transition Pathway of the Signaling Protein Nitrogen Regulatory Protein C
- Author
-
Ming Lei, Janice Velos, Martin Karplus, Alexandra K. Gardino, Alexsandr Kivenson, and Dorothee Kern
- Subjects
Models, Molecular ,Protein Conformation ,Chemistry ,Hydrogen bond ,Escherichia coli Proteins ,PII Nitrogen Regulatory Proteins ,Molecular Sequence Data ,Hydrogen Bonding ,Article ,Molecular dynamics ,Protein structure ,Biochemistry ,Structural Biology ,Biophysics ,Phosphorylation ,Computer Simulation ,Pii nitrogen regulatory proteins ,Signal transduction ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Transcription factor ,Function (biology) ,Signal Transduction ,Transcription Factors - Abstract
Recent advances in experimental methods provide increasing evidence that proteins sample the conformational substates that are important for function in the absence of their ligands. An example is the receiver domain of nitrogen regulatory protein C, a member of the phosphorylation-mediated signaling family of "two-component systems." The receiver domain of nitrogen regulatory protein C samples both inactive conformation and the active conformation before phosphorylation. Here we determine a possible pathway of interconversion between the active state and the inactive state by targeted molecular dynamics simulations and quasi-harmonic analysis; these methods are used because the experimental conversion rate is in the high microsecond range, longer than those that are easily accessible to atomistic molecular dynamics simulations. The calculated pathway is found to be composed of four consecutive stages described by different progress variables. The lowest quasi-harmonic principal components from unbiased molecular dynamics simulations on the active state correspond to the first stage, but not to the subsequent stages of the transition. The targeted molecular dynamics pathway suggests that several transient nonnative hydrogen bonds may facilitate the transition.
- Published
- 2009
- Full Text
- View/download PDF
35. Molecular Mechanism of Pin1-Tau Recognition and Catalysis
- Author
-
Timo Eichner, Steffen Kutter, Wladimir Labeikovsky, Dorothee Kern, and Vanessa Buosi
- Subjects
0301 basic medicine ,Magnetic Resonance Spectroscopy ,tau Proteins ,Calorimetry ,Dephosphorylation ,WW domain ,03 medical and health sciences ,Structural Biology ,Scattering, Small Angle ,medicine ,Humans ,Molecular Biology ,biology ,Chemistry ,Binding protein ,Neurodegeneration ,Isothermal titration calorimetry ,Protein phosphatase 2 ,medicine.disease ,NIMA-Interacting Peptidylprolyl Isomerase ,030104 developmental biology ,Biochemistry ,Biophysics ,PIN1 ,biology.protein ,Phosphorylation ,Protein Processing, Post-Translational ,Protein Binding - Abstract
Human peptidyl-prolyl isomerase (PPIase) Pin1 plays key roles in developmental processes, cell proliferation, and neuronal function. Extensive phosphorylation of the microtubule binding protein tau has been implicated in neurodegeneration and Alzheimer's disease. For the past 15years, these two players have been the focus of an enormous research effort to unravel the biological relevance of their interplay in health and disease, resulting in a series of proposed molecular mechanism of how Pin1 catalysis of tau results in biological phenotypes. Our results presented here refute these mechanisms of Pin1 action. Using NMR, isothermal calorimetry (ITC), and small angle x-ray scattering (SAXS), we dissect binding and catalysis on multiple phosphorylated tau with particular emphasis toward the Alzheimer's associated AT180 tau epitope containing phosphorylated THR231 and SER235. We find that phosphorylated (p-) SER235-PRO, but not pTHR231-PRO, is exclusively catalyzed by full-length Pin1 and isolated PPIase domain. Importantly, site-specific measurements of Pin1-catalysis of CDK2/CycA-phosphorylated full-length tau reveal a number of sites that are catalyzed simultaneously with different efficiencies. Furthermore, we show that the turnover efficiency at pSER235 by Pin1 is independent of both the WW domain and phosphorylation on THR231. Our mechanistic results on site-specific binding and catalysis together with the lack of an increase of dephosphorylation rates by PP2A counter a series of previously published models for the role of Pin1 catalysis of tau in Alzheimer's disease. Together, our data reemphasize the complicated scenario between binding and catalysis of multiple phosphorylated tau by Pin1 and the need for directly linking biological phenotypes and residue-specific turnover in Pin1 substrates.
- Published
- 2015
36. Linkage between dynamics and catalysis in a thermophilic-mesophilic enzyme pair
- Author
-
Dorothee Kern, Georgia Hadjipavlou, Vu Hong Thai, Elan Z. Eisenmesser, Magnus Wolf-Watz, and Katherine A. Henzler-Wildman
- Subjects
chemistry.chemical_classification ,Chemistry ,Protein dynamics ,Thermophile ,Adenylate Kinase ,Adenylate kinase ,Chemical reaction ,Catalysis ,Enzyme structure ,Enzyme ,Biochemistry ,Structural Biology ,Biophysics ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Mesophile - Abstract
A fundamental question is how enzymes can accelerate chemical reactions. Catalysis is not only defined by actual chemical steps, but also by enzyme structure and dynamics. To investigate the role of protein dynamics in enzymatic turnover, we measured residue-specific protein dynamics in hyperthermophilic and mesophilic homologs of adenylate kinase during catalysis. A dynamic process, the opening of the nucleotide-binding lids, was found to be rate-limiting for both enzymes as measured by NMR relaxation. Moreover, we found that the reduced catalytic activity of the hyperthermophilic enzyme at ambient temperatures is caused solely by a slower lid-opening rate. This comparative and quantitative study of activity, structure and dynamics revealed a close link between protein dynamics and catalytic turnover.
- Published
- 2004
- Full Text
- View/download PDF
37. The role of dynamics in allosteric regulation
- Author
-
Erik R. P. Zuiderweg and Dorothee Kern
- Subjects
Models, Molecular ,Protein Folding ,Binding Sites ,Protein Conformation ,Chemistry ,Allosteric regulation ,Proteins ,Stereoisomerism ,Kinetics ,Motion ,Protein structure ,Models, Chemical ,Structural Biology ,Computational chemistry ,Protein folding ,Molecular Biology ,Neuroscience ,Protein Binding - Abstract
The biomolecular conformational changes often associated with allostery are, by definition, dynamic processes. Recent publications have disclosed the role of pre-existing equilibria of conformational substates in this process. In addition, the role of dynamics as an entropic carrier of free energy of allostery has been investigated. Recent work thus shows that dynamics is pivotal to allostery, and that it constitutes much more than just the move from the 'T'-state to the 'R'-state. Emerging computational studies have described the actual pathways of allosteric change.
- Published
- 2003
- Full Text
- View/download PDF
38. The NMR Solution Structure of BeF3−-Activated Spo0F Reveals the Conformational Switch in a Phosphorelay System
- Author
-
Dorothee Kern, Ho S. Cho, Alexandra K. Gardino, Brian F. Volkman, Seok-Yong Lee, and David E. Wemmer
- Subjects
Models, Molecular ,Protein Conformation ,Turn (biochemistry) ,Fluorides ,Bacterial Proteins ,Structural Biology ,Transferase ,Histidine ,Phosphorylation ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Aspartic Acid ,Chemistry ,Phosphotransferases ,fungi ,Histidine kinase ,Nuclear magnetic resonance spectroscopy ,Response regulator ,Biochemistry ,Helix ,Biophysics ,Beryllium ,Signal transduction ,Bacillus subtilis ,Signal Transduction - Abstract
Two-component systems, which are comprised of a single histidine-aspartate phosphotransfer module, are the dominant signaling pathways in bacteria and have recently been identified in several eukaryotic organisms as well. A tandem connection of two or more histidine-aspartate motifs forms complex phosphorelays. While response regulators from simple two-component systems have been characterized structurally in their inactive and active forms, we address here the question of whether a response regulator from a phosphorelay has a distinct structural basis of activation. We report the NMR solution structure of BeF3−-activated Spo0F, the first structure of a response regulator from a phosphorelay in its activated state. Conformational changes were found in regions previously identified to change in simple two-component systems. In addition, a downward shift by half a helical turn in helix 1, located on the opposite side of the common activation surface, was observed as a consequence of BeF3− activation. Conformational changes in helix 1 can be rationalized by the distinct function of phosphoryl transfer to the second histidine kinase, Spo0B, because helix 1 is known to interact directly with Spo0B and the phosphatase RapB. The identification of structural rearrangements in Spo0F supports the hypothesis of a pre-existing equilibrium between the inactive and active state prior to phosphorylation that was suggested on the basis of previous NMR dynamics studies on Spo0F. A shift of a pre-existing equilibrium is likely a general feature of response regulators.
- Published
- 2003
- Full Text
- View/download PDF
39. Regulation of Kinases: 1 Billion Years of Evolution
- Author
-
Christine D. Wilson, Sarita Biswas, Roman V. Agafonov, and Dorothee Kern
- Subjects
ABL ,Common descent ,Mechanism (biology) ,Kinase ,Biophysics ,Phosphorylation ,Protein phosphorylation ,Biology ,Tyrosine ,Cell biology ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Protein phosphorylation is an essential regulatory mechanism that affects all aspect of cellular life from division and growth to aging and death. Misregulation of the signaling cascades leads to severe detrimental effects, and in humans often associated with cancer and other diseases. Phosphorylation is performed by a class of protein called kinases. Activation and deactivation of kinases is normally under tight control and is regulated via different mechanisms that are incredibly complex. In this work we combine phylogenetic resurrection techniques with biophysical and chemical approaches to analyze the regulatory mechanisms of modern tyrosine oncokinases Src and Abl, their common ancestor and the common ancestors between several other families of tyrosin kinases. Our results show how the regulatory elements appeared and developed throughout the evolution enabling selective regulation of complex modern cascades.
- Published
- 2015
- Full Text
- View/download PDF
40. Structural properties of the histidine-containing loop in HIV-1 RNase H
- Author
-
Dorothee Kern, Gunther Kern, Jeffrey G. Pelton, and Susan Marqusee
- Subjects
Protein Denaturation ,Protein Conformation ,RNase P ,Stereochemistry ,Ribonuclease H ,Biophysics ,Biochemistry ,Structure-Activity Relationship ,Protein structure ,Humans ,Urea ,Structure–activity relationship ,Histidine ,RNase H ,Nuclear Magnetic Resonance, Biomolecular ,biology ,Chemistry ,Organic Chemistry ,Hydrogen-Ion Concentration ,Crystallography ,Helix ,HIV-1 ,biology.protein ,Chemical stability ,Protein folding - Abstract
The isolated HIV-1 RNase H domain is inactive. This inactivity has been linked to the lack of structure in the C-terminus of the isolated domain. Thermodynamic stability experiments on the RNase H domain as well as a deletion mutant lacking the C-terminal helix have implied that this region is structured. His539 residing in a loop preceding the C-terminal helix was studied by NMR to determine the stability and conformational properties of this region. The stability of the structural environment of His539 matches that of the entire RNase H domain. Furthermore, His539 is locked into a defined tautometric state in the folded protein and its pK(a) is shifted compared to a freely accessible His, suggesting that this region is structured. The data support the view that the overall dynamics rather than the lack of structure in a small portion of the protein render activity of the isolated HIV-1 RNase H.
- Published
- 2002
- Full Text
- View/download PDF
41. Catalysis of cis/trans isomerization in native HIV-1 capsid by human cyclophilin A
- Author
-
Susan Sondej Pochapsky, Dorothee Kern, Daryl A. Bosco, Wesley I. Sundquist, and Elan Z. Eisenmesser
- Subjects
Models, Molecular ,Protein Folding ,Magnetic Resonance Spectroscopy ,Protein Conformation ,Stereochemistry ,Genetic Vectors ,Cypa ,Isomerase ,Plasma protein binding ,Crystallography, X-Ray ,Catalysis ,Cyclophilin A ,Capsid ,Protein structure ,Humans ,Peptide bond ,Multidisciplinary ,Virulence ,biology ,Chemistry ,food and beverages ,Stereoisomerism ,Biological Sciences ,biology.organism_classification ,Cis trans isomerization ,Protein Structure, Tertiary ,Kinetics ,Models, Chemical ,HIV-1 ,Protein folding ,Peptides ,Protein Binding - Abstract
Packaging of cyclophilin A (CypA) into HIV-1 virions is essential for efficient replication; however, the reason for this is unknown. Incorporation is mediated through binding to the Gly-89–Pro-90 peptide bond of the N-terminal domain of HIV-1 capsid (CA N ). Despite the fact that CypA is a peptidyl-prolyl cis/trans isomerase, catalytic activity on CA N has not been observed previously. We show here, using NMR exchange spectroscopy, that CypA does not only bind to CA N but also catalyzes efficiently the cis/trans isomerization of the Gly-89–Pro-90 peptide bond. In addition, conformational changes in CA N distal to the CypA binding loop are observed on CypA binding and catalysis. The results provide experimental evidence for efficient CypA catalysis on a natively folded and biologically relevant protein substrate.
- Published
- 2002
- Full Text
- View/download PDF
42. Second Harmonic Generation as a Method to Identify and Screen for Allosteric Modulators of Protein Targets
- Author
-
Elizabeth Donohue Vo, Roman V. Agafonov, Gabriel Mercado, Katelyn Connell, Dorothee Kern, Joshua Salafsky, Frank McCormick, and Tad George
- Subjects
Protein function ,biology ,Allosteric enzyme ,Drug target ,Allosteric regulation ,Biophysics ,biology.protein ,Active site ,Computational biology ,Binding site ,Molecular biology - Abstract
Proteins populate a landscape of conformations that changes upon binding native ligands or drugs, altering protein function. Different conformations reveal distinct potential drug target sites, including allosteric sites distal to the active site of the protein. It has been historically difficult to identify allosteric modulators early in the screening process since the structural methods required to reveal binding sites are relatively low throughput.
- Published
- 2017
- Full Text
- View/download PDF
43. Molecular mechanism of Aurora A kinase autophosphorylation and its allosteric activation by TPX2
- Author
-
Young-Jin Cho, Vanessa Buosi, Francesco Pontiggia, Dorothee Kern, Adelajda Zorba, Nadja Kern, and Steffen Kutter
- Subjects
Models, Molecular ,QH301-705.5 ,kinase ,Protein Conformation ,Science ,Allosteric regulation ,Aurora A kinase ,mechanism ,Cell Cycle Proteins ,Biology ,Crystallography, X-Ray ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,Substrate Specificity ,Allosteric Regulation ,Humans ,Biology (General) ,Phosphorylation ,Mitosis ,Aurora Kinase A ,General Immunology and Microbiology ,Kinase ,Protein Stability ,General Neuroscience ,Autophosphorylation ,E. coli ,Nuclear Proteins ,General Medicine ,Biophysics and Structural Biology ,3. Good health ,Cell biology ,Spindle apparatus ,Protein Structure, Tertiary ,Solutions ,Kinetics ,Phosphothreonine ,Centrosome ,Biocatalysis ,Medicine ,activation ,Mutant Proteins ,Protein Multimerization ,Microtubule-Associated Proteins ,Research Article ,Protein Binding - Abstract
We elucidate the molecular mechanisms of two distinct activation strategies (autophosphorylation and TPX2-mediated activation) in human Aurora A kinase. Classic allosteric activation is in play where either activation loop phosphorylation or TPX2 binding to a conserved hydrophobic groove shifts the equilibrium far towards the active conformation. We resolve the controversy about the mechanism of autophosphorylation by demonstrating intermolecular autophosphorylation in a long-lived dimer by combining X-ray crystallography with functional assays. We then address the allosteric activation by TPX2 through activity assays and the crystal structure of a domain-swapped dimer of dephosphorylated Aurora A and TPX21−25. While autophosphorylation is the key regulatory mechanism in the centrosomes in the early stages of mitosis, allosteric activation by TPX2 of dephosphorylated Aurora A could be at play in the spindle microtubules. The mechanistic insights into autophosphorylation and allosteric activation by TPX2 binding proposed here, may have implications for understanding regulation of other protein kinases. DOI: http://dx.doi.org/10.7554/eLife.02667.001, eLife digest The kinase, Aurora A, is a human protein that is needed for cells to divide normally. Kinases are enzymes that control other proteins by adding phosphate groups to these proteins; however, like other kinases, Aurora A must first be activated or ‘switched on’ before it can do this. Aurora A kinase can be switched on in two ways: by having a phosphate group added to its ‘activation loop’; or by binding to another protein called TPX2. Also like other kinases, Aurora A can self-activate, but the details of this process are not understood. Does a single Aurora A kinase add a phosphate group to its own activation loop, or does one Aurora A kinase activate a second? Furthermore, it is not clear how binding to TPX2 can activate an Aurora A kinase without adding a phosphate group to the activation loop. Zorba, Buosi et al. now show that Aurora A kinases that have been activated in different ways—via the addition of a phosphate group or binding to TPX2—are equally good at adding phosphate groups to other proteins. Zorba, Buosi et al. also worked out the three-dimensional shapes of the kinases activated in these two ways—since many proteins change shape when they are switched on—and found that they were also the same. Finally, it was shown that self-activation involves two Aurora A kinases binding to each other, and one kinase adding a phosphate group to the other, rather than a single kinase adding a phosphate group to itself. Since other protein kinases can be activated in similar ways to Aurora A, the findings of Zorba, Buosi et al. might also help us to understand how other protein kinases can be switched ‘on’ or ‘off’. And, as mutations in Aurora A have been linked to the development of cancer, uncovering how this kinase is controlled could help efforts to design new drugs to treat this disease. DOI: http://dx.doi.org/10.7554/eLife.02667.002
- Published
- 2014
44. Author response: Molecular mechanism of Aurora A kinase autophosphorylation and its allosteric activation by TPX2
- Author
-
Young-Jin Cho, Adelajda Zorba, Nadja Kern, Francesco Pontiggia, Steffen Kutter, Dorothee Kern, and Vanessa Buosi
- Subjects
Chemistry ,Allosteric regulation ,Autophosphorylation ,Molecular mechanism ,Aurora A kinase ,Cell biology - Published
- 2014
- Full Text
- View/download PDF
45. Structure of a transiently phosphorylated switch in bacterial signal transduction
- Author
-
Sydney Kustu, David E. Wemmer, Dorothee Kern, Brian F. Volkman, Peter Luginbuhl, and Michael J. Nohaile
- Subjects
Models, Molecular ,Conformational change ,Binding Sites ,Magnetic Resonance Spectroscopy ,Multidisciplinary ,Protein Conformation ,Stereochemistry ,PII Nitrogen Regulatory Proteins ,Autophosphorylation ,Active site ,Biology ,DNA-Binding Proteins ,Dephosphorylation ,Protein structure ,Bacterial Proteins ,Trans-Activators ,biology.protein ,Biophysics ,Phosphorylation ,Pii nitrogen regulatory proteins ,Signal transduction ,Signal Transduction ,Transcription Factors - Abstract
Receiver domains are the dominant molecular switches in bacterial signalling. Although several structures of non-phosphorylated receiver domains have been reported, a detailed structural understanding of the activation arising from phosphorylation has been impeded by the very short half-lives of the aspartylphosphate linkages. Here we present the first structure of a receiver domain in its active state, the phosphorylated receiver domain of the bacterial enhancer-binding protein NtrC (nitrogen regulatory protein C). Nuclear magnetic resonance spectra were taken during steady-state autophosphorylation/dephosphorylation, and three-dimensional spectra from multiple samples were combined. Phosphorylation induces a large conformational change involving a displacement of beta-strands 4 and 5 and alpha-helices 3 and 4 away from the active site, a register shift and an axial rotation in helix 4. This creates an exposed hydrophobic surface that is likely to transmit the signal to the transcriptional activation domain.
- Published
- 1999
- Full Text
- View/download PDF
46. Evidence against the ‘Y-T coupling’ mechanism of activation in the response regulator NtrC
- Author
-
Dorothee Kern, Michael W. Clarkson, Janice Villali, Michael F. Hagan, and Francesco Pontiggia
- Subjects
Magnetic Resonance Spectroscopy ,Chemistry ,Stereochemistry ,Protein dynamics ,PII Nitrogen Regulatory Proteins ,Allosteric regulation ,Article ,Coupling (electronics) ,Response regulator ,Structural Biology ,Phosphorylation ,Pii nitrogen regulatory proteins ,Computer Simulation ,Tyrosine ,Molecular Biology ,Conformational isomerism ,Allosteric Site - Abstract
The dominant theory on the mechanism of response regulators activation in two-component bacterial signaling systems is the "Y-T coupling" mechanism, wherein the χ1 rotameric state of a highly conserved aromatic residue correlates with the activation of the protein via structural rearrangements coupled to a conserved tyrosine. In this paper, we present evidence that, in the receiver domain of the response regulator nitrogen regulatory protein C (NtrC(R)), the interconversion of this tyrosine (Y101) between its rotameric states is actually faster than the rate of inactive/active conversion and is not correlated to the activation process. Data gathered from NMR relaxation dispersion experiments show that a subset of residues surrounding the conserved tyrosine sense a process that is occurring at a faster rate than the inactive/active conformational transition. We show that this process is related to χ1 rotamer exchange of Y101 and that mutation of this aromatic residue to a leucine eliminated this second faster process without affecting activation. Computational simulations of NtrC(R) in its active conformation further demonstrate that the rotameric state of Y101 is uncorrelated with the global conformational transition during activation. Moreover, the tyrosine does not appear to be involved in the stabilization of the active form upon phosphorylation and is not essential in propagating the signal downstream for ATPase activity of the central domain. Our data provide experimental evidence against the generally accepted "Y-T coupling" mechanism of activation in NtrC(R).
- Published
- 2014
47. Structural and functional analyses of activating amino acid substitutions in the receiver domain of NtrC: Evidence for an activating surface
- Author
-
Michael J. Nohaile, Kenneth M. Stedman, Dorothee Kern, David E. Wemmer, and Sydney Kustu
- Subjects
DNA, Bacterial ,Models, Molecular ,Transcriptional Activation ,Magnetic Resonance Spectroscopy ,Protein Conformation ,PII Nitrogen Regulatory Proteins ,Recombinant Fusion Proteins ,Mutant ,Gene Expression ,Methyl-Accepting Chemotaxis Proteins ,Protein Structure, Secondary ,Bacterial Proteins ,Structural Biology ,Transcription (biology) ,Escherichia coli ,Magnesium ,Cloning, Molecular ,Phosphorylation ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,biology ,Chemistry ,Escherichia coli Proteins ,Membrane Proteins ,Active site ,DNA-Directed RNA Polymerases ,Amino acid ,DNA-Binding Proteins ,Biochemistry ,Mutagenesis ,Trans-Activators ,biology.protein ,Biophysics ,Signal transduction ,Two-dimensional nuclear magnetic resonance spectroscopy ,Heteronuclear single quantum coherence spectroscopy ,Signal Transduction ,Transcription Factors - Abstract
The bacterial enhancer-binding protein NtrC activates transcription when phosphorylated on aspartate 54 in its amino (N)-terminal regulatory domain or when altered by constitutively activating amino acid substitutions. The N-terminal domain of NtrC, which acts positively on the remainder of the protein, is homologous to a large family of signal transduction domains called receiver domains. Phosphorylation of an aspartate residue in a receiver domain modulates the function of a downstream target, but the accompanying structural changes are not clear. In the present work we examine structural and functional differences between the wild-type receiver domain of NtrC and mutant forms carrying constitutively activating substitutions. Combinations of such substitutions resulted in both increased structural changes in the N-terminal domain, monitored by NMR chemical shift differences, and increased transcriptional activation by the full-length protein. Structural changes caused by substitutions outside the active site (D86N and A89T) were not only local but extended over a substantial portion of the N-terminal domain including the region from α-helix 3 to β-strand 5 (“3445 face”) and propagating to the active site. Interestingly, the activating substitution of glutamate for aspartate at the site of phosphorylation (D54E) also triggered structural changes in the 3445 face. Thus, the active site and the 3445 face appear to interact. Implications with respect to how phosphorylation may affect the structure of receiver domains and how structural changes may be communicated to the remainder of NtrC are discussed.
- Published
- 1997
- Full Text
- View/download PDF
48. Rotational Barriers ofcis/transIsomerization of Proline Analogues and Their Catalysis by Cyclophilin
- Author
-
Mike Schutkowski, Dorothee Kern, and Torbjörn Drakenberg
- Subjects
chemistry.chemical_classification ,Stereochemistry ,General Chemistry ,Biochemistry ,Catalysis ,Cis trans isomerization ,Amino acid ,Hydroxyproline ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,chemistry ,Proline ,Isomerization ,Cis–trans isomerism ,Cyclophilin - Abstract
The rotational barriers for cis/trans isomerization of different proline analogues have been investigated by dynamic 1H NMR spectroscopy. To this end the analogues (S)-azetidine-2-carboxylic acid (Aze), (S)-piperidine-2-carboxylic acid (Pip), (R)-thiazolidine-4-carboxylic acid (4-Thz), (4R)-2-methylthiazolidine-4-carboxylic acid (2Me4-Thz), (R)-thiazolidine-2-carboxylic acid (2-Thz), (S)-oxazolidine-4-carboxylic acid (4-Oxa), (4S,5R)-5-methyloxazolidine-4-carboxylic acid (5Me4-Oxa), and (2S,4R)-4-hydroxypyrrolidine-2-carboxylic acid (Hyp) and several N-alkylated amino acids were incorporated into the sequences Ala-Yaa-(4-)nitroanilide and Ala-Gly-Yaa-Phe-(4-)nitroanilide. NMR line-shape analyses of various cis and trans proton signals of these peptides were performed at different temperatures, and the rate constants of cis/trans isomerization were fitted to the Eyring equation. The rotational barriers of all cyclic proline analogues except hydroxyproline were found to be lower than that of proline by abou...
- Published
- 1997
- Full Text
- View/download PDF
49. How Thiamine Diphosphate Is Activated in Enzymes
- Author
-
Holger Neef, Gerhard Hübner, Dorothee Kern, Kai Tittmann, Christer Wikner, Margrit Killenberg-Jabs, Gunter Schneider, and Gunther Kern
- Subjects
Magnetic Resonance Spectroscopy ,Pyrimidine ,Stereochemistry ,Glutamic Acid ,Transketolase ,Catalysis ,Benzoylformate decarboxylase ,Cofactor ,chemistry.chemical_compound ,Deprotonation ,Allosteric Regulation ,Pyruvates ,chemistry.chemical_classification ,Binding Sites ,Multidisciplinary ,biology ,Hydrogen-Ion Concentration ,Deuterium ,Enzyme Activation ,Kinetics ,Enzyme ,chemistry ,Mutagenesis, Site-Directed ,biology.protein ,Thermodynamics ,Thiamine ,Protons ,Thiamine Pyrophosphate ,Pyruvate Decarboxylase ,Pyruvate decarboxylase - Abstract
The controversial question of how thiamine diphosphate, the biologically active form of vitamin B 1 , is activated in different enzymes has been addressed. Activation of the coenzyme was studied by measuring thermodynamics and kinetics of deprotonation at the carbon in the 2-position (C2) of thiamine diphosphate in the enzymes pyruvate decarboxylase and transketolase by use of nuclear magnetic resonance spectroscopy, proton/deuterium exchange, coenzyme analogs, and site-specific mutant enzymes. Interaction of a glutamate with the nitrogen in the 1′-position in the pyrimidine ring activated the 4′-amino group to act as an efficient proton acceptor for the C2 proton. The protein component accelerated the deprotonation of the C2 atom by several orders of magnitude, beyond the rate of the overall enzyme reaction. Therefore, the earlier proposed concerted mechanism or stabilization of a C2 carbanion can be excluded.
- Published
- 1997
- Full Text
- View/download PDF
50. A Highly Conserved Cysteine of Neuronal Calcium-sensing Proteins Controls Cooperative Binding of Ca2+ to Recoverin*
- Author
-
Kalyan S. Chakrabarti, Daniel D. Oprian, Ramasamy P. Kumar, Matthew J. Ranaghan, Dorothee Kern, and Vanessa Buosi
- Subjects
Models, Molecular ,genetic structures ,Amino Acid Motifs ,Cooperativity ,Plasma protein binding ,Biochemistry ,behavioral disciplines and activities ,Recoverin ,Calcium-binding protein ,mental disorders ,Protein myristoylation ,Amino Acid Sequence ,Cysteine ,Molecular Biology ,Conserved Sequence ,Myristoylation ,biology ,Cooperative binding ,Cell Biology ,eye diseases ,Rhodopsin kinase ,Mutagenesis ,Mutation ,Biophysics ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Calcium ,sense organs ,Oxidation-Reduction ,Signal Transduction ,Protein Binding - Abstract
Recoverin, a 23-kDa Ca(2+)-binding protein of the neuronal calcium sensing (NCS) family, inhibits rhodopsin kinase, a Ser/Thr kinase responsible for termination of photoactivated rhodopsin in rod photoreceptor cells. Recoverin has two functional EF hands and a myristoylated N terminus. The myristoyl chain imparts cooperativity to the Ca(2+)-binding sites through an allosteric mechanism involving a conformational equilibrium between R and T states of the protein. Ca(2+) binds preferentially to the R state; the myristoyl chain binds preferentially to the T state. In the absence of myristoylation, the R state predominates, and consequently, binding of Ca(2+) to the non-myristoylated protein is not cooperative. We show here that a mutation, C39A, of a highly conserved Cys residue among NCS proteins, increases the apparent cooperativity for binding of Ca(2+) to non-myristoylated recoverin. The binding data can be explained by an effect on the T/R equilibrium to favor the T state without affecting the intrinsic binding constants for the two Ca(2+) sites.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.