73 results on '"Biondi RM"'
Search Results
2. Molecular insights into the regulatory landscape of PKC-related kinase-2 (PRK2/PKN2) using targeted small compounds.
- Author
-
Gross LZF, Winkel AF, Galceran F, Schulze JO, Fröhner W, Cämmerer S, Zeuzem S, Engel M, Leroux AE, and Biondi RM
- Subjects
- Humans, Allosteric Regulation, Catalytic Domain, Molecular Docking Simulation, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases chemistry, 3-Phosphoinositide-Dependent Protein Kinases metabolism, 3-Phosphoinositide-Dependent Protein Kinases genetics, 3-Phosphoinositide-Dependent Protein Kinases chemistry, Protein Binding, Protein Kinase C metabolism, Protein Kinase C genetics, Protein Kinase C chemistry, Pyruvate Dehydrogenase Acetyl-Transferring Kinase metabolism, Pyruvate Dehydrogenase Acetyl-Transferring Kinase genetics
- Abstract
The PKC-related kinases (PRKs, also termed PKNs) are important in cell migration, cancer, hepatitis C infection, and nutrient sensing. They belong to a group of protein kinases called AGC kinases that share common features like a C-terminal extension to the catalytic domain comprising a hydrophobic motif. PRKs are regulated by N-terminal domains, a pseudosubstrate sequence, Rho-binding domains, and a C2 domain involved in inhibition and dimerization, while Rho and lipids are activators. We investigated the allosteric regulation of PRK2 and its interaction with its upstream kinase PDK1 using a chemical biology approach. We confirmed the phosphoinositide-dependent protein kinase 1 (PDK1)-interacting fragment (PIF)-mediated docking interaction of PRK2 with PDK1 and showed that this interaction can be modulated allosterically. We showed that the polypeptide PIFtide and a small compound binding to the PIF-pocket of PRK2 were allosteric activators, by displacing the pseudosubstrate PKL region from the active site. In addition, a small compound binding to the PIF-pocket allosterically inhibited the catalytic activity of PRK2. Together, we confirmed the docking interaction and allostery between PRK2 and PDK1 and described an allosteric communication between the PIF-pocket and the active site of PRK2, both modulating the conformation of the ATP-binding site and the pseudosubstrate PKL-binding site. Our study highlights the allosteric modulation of the activity and the conformation of PRK2 in addition to the existence of at least two different complexes between PRK2 and its upstream kinase PDK1. Finally, the study highlights the potential for developing allosteric drugs to modulate PRK2 kinase conformations and catalytic activity., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. The PB1 and the ZZ domain of the autophagy receptor p62/SQSTM1 regulate the interaction of p62/SQSTM1 with the autophagosome protein LC3B.
- Author
-
Alcober-Boquet L, Zang T, Pietsch L, Suess E, Hartmann M, Proschak E, Gross LZF, Sacerdoti M, Zeuzem S, Rogov VV, Leroux AE, Piiper A, and Biondi RM
- Subjects
- Sequestosome-1 Protein genetics, Sequestosome-1 Protein metabolism, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Autophagy genetics, Autophagosomes metabolism, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism
- Abstract
Autophagy is a highly conserved cellular process that allows degradation of large macromolecules. p62/SQSTM1 is a key adaptor protein that interacts both with material to be degraded and with LC3 at the autophagosome, enabling degradation of cargos such as protein aggregates, lipid droplets and damaged organelles by selective autophagy. Dysregulation of autophagy contributes to the pathogenesis of many diseases. In this study, we investigated if the interaction of p62/SQSTM1 with LC3B could be regulated. We purified full-length p62/SQSTM1 and established an in vitro assay that measures the interaction with LC3B. We used the assay to determine the role of the different domains of p62/SQSTM1 in the interaction with LC3B. We identified a mechanism of regulation of p62/SQSTM1 where the ZZ and the PB1 domains regulate the exposure of the LIR-sequence to enable or inhibit the interaction with LC3B. A mutation to mimic the phosphorylation of a site on the ZZ domain leads to increased interaction with LC3B. Also, a small compound that binds to the ZZ domain enhances interaction with LC3B. Dysregulation of these mechanisms in p62/SQSTM1 could have implications for diseases where autophagy is affected. In conclusion, our study highlights the regulated nature of p62/SQSTM1 and its ability to modulate the interaction with LC3B through a LIR-sequence Accessibility Mechanism (LAM). Furthermore, our findings suggest the potential for pharmacological modulation of the exposure of LIR, paving the way for future therapeutic strategies., (© 2023 The Authors. Protein Science published by Wiley Periodicals LLC on behalf of The Protein Society.)
- Published
- 2024
- Full Text
- View/download PDF
4. The choreography of protein kinase PDK1 and its diverse substrate dance partners.
- Author
-
Leroux AE and Biondi RM
- Subjects
- Humans, Binding Sites, Phosphatidylinositol 3-Kinase, Phosphorylation, Proto-Oncogene Proteins c-akt, Pyruvate Dehydrogenase Acetyl-Transferring Kinase metabolism
- Abstract
The protein kinase PDK1 phosphorylates at least 24 distinct substrates, all of which belong to the AGC protein kinase group. Some substrates, such as conventional PKCs, undergo phosphorylation by PDK1 during their synthesis and subsequently get activated by DAG and Calcium. On the other hand, other substrates, including members of the Akt/PKB, S6K, SGK, and RSK families, undergo phosphorylation and activation downstream of PI3-kinase signaling. This review presents two accepted molecular mechanisms that determine the precise and timely phosphorylation of different substrates by PDK1. The first mechanism involves the colocalization of PDK1 with Akt/PKB in the presence of PIP3. The second mechanism involves the regulated docking interaction between the hydrophobic motif (HM) of substrates and the PIF-pocket of PDK1. This interaction, in trans, is equivalent to the molecular mechanism that governs the activity of AGC kinases through their HMs intramolecularly. PDK1 has been instrumental in illustrating the bi-directional allosteric communication between the PIF-pocket and the ATP-binding site and the potential of the system for drug discovery. PDK1's interaction with substrates is not solely regulated by the substrates themselves. Recent research indicates that full-length PDK1 can adopt various conformations based on the positioning of the PH domain relative to the catalytic domain. These distinct conformations of full-length PDK1 can influence the interaction and phosphorylation of substrates. Finally, we critically discuss recent findings proposing that PIP3 can directly regulate the activity of PDK1, which contradicts extensive in vitro and in vivo studies conducted over the years., (© 2023 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2023
- Full Text
- View/download PDF
5. Modulation of the substrate specificity of the kinase PDK1 by distinct conformations of the full-length protein.
- Author
-
Sacerdoti M, Gross LZF, Riley AM, Zehnder K, Ghode A, Klinke S, Anand GS, Paris K, Winkel A, Herbrand AK, Godage HY, Cozier GE, Süß E, Schulze JO, Pastor-Flores D, Bollini M, Cappellari MV, Svergun D, Gräwert MA, Aramendia PF, Leroux AE, Potter BVL, Camacho CJ, and Biondi RM
- Subjects
- Animals, Substrate Specificity, Phosphorylation, Catalytic Domain, Dimerization, Polyphosphates, Mammals
- Abstract
The activation of at least 23 different mammalian kinases requires the phosphorylation of their hydrophobic motifs by the kinase PDK1. A linker connects the phosphoinositide-binding PH domain to the catalytic domain, which contains a docking site for substrates called the PIF pocket. Here, we used a chemical biology approach to show that PDK1 existed in equilibrium between at least three distinct conformations with differing substrate specificities. The inositol polyphosphate derivative HYG8 bound to the PH domain and disrupted PDK1 dimerization by stabilizing a monomeric conformation in which the PH domain associated with the catalytic domain and the PIF pocket was accessible. In the absence of lipids, HYG8 potently inhibited the phosphorylation of Akt (also termed PKB) but did not affect the intrinsic activity of PDK1 or the phosphorylation of SGK, which requires docking to the PIF pocket. In contrast, the small-molecule valsartan bound to the PIF pocket and stabilized a second distinct monomeric conformation. Our study reveals dynamic conformations of full-length PDK1 in which the location of the linker and the PH domain relative to the catalytic domain determines the selective phosphorylation of PDK1 substrates. The study further suggests new approaches for the design of drugs to selectively modulate signaling downstream of PDK1.
- Published
- 2023
- Full Text
- View/download PDF
6. Characterization of p38α autophosphorylation inhibitors that target the non-canonical activation pathway.
- Author
-
González L, Díaz L, Pous J, Baginski B, Duran-Corbera A, Scarpa M, Brun-Heath I, Igea A, Martin-Malpartida P, Ruiz L, Pallara C, Esguerra M, Colizzi F, Mayor-Ruiz C, Biondi RM, Soliva R, Macias MJ, Orozco M, and Nebreda AR
- Subjects
- Humans, Phosphorylation, Inflammation, Signal Transduction
- Abstract
p38α is a versatile protein kinase that can control numerous processes and plays important roles in the cellular responses to stress. Dysregulation of p38α signaling has been linked to several diseases including inflammation, immune disorders and cancer, suggesting that targeting p38α could be therapeutically beneficial. Over the last two decades, numerous p38α inhibitors have been developed, which showed promising effects in pre-clinical studies but results from clinical trials have been disappointing, fueling the interest in the generation of alternative mechanisms of p38α modulation. Here, we report the in silico identification of compounds that we refer to as non-canonical p38α inhibitors (NC-p38i). By combining biochemical and structural analyses, we show that NC-p38i efficiently inhibit p38α autophosphorylation but weakly affect the activity of the canonical pathway. Our results demonstrate how the structural plasticity of p38α can be leveraged to develop therapeutic opportunities targeting a subset of the functions regulated by this pathway., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
7. A Tetratricopeptide Repeat Scaffold Couples Signal Detection to OdhI Phosphorylation in Metabolic Control by the Protein Kinase PknG.
- Author
-
Lisa MN, Sogues A, Barilone N, Baumgart M, Gil M, Graña M, Durán R, Biondi RM, Bellinzoni M, Bott M, and Alzari PM
- Subjects
- Amino Acids metabolism, Bacterial Proteins genetics, Corynebacterium glutamicum genetics, Corynebacterium glutamicum metabolism, Gene Expression Regulation, Bacterial, Glutamic Acid metabolism, Mycobacterium tuberculosis genetics, Mycobacterium tuberculosis metabolism, Phosphorylation, Protein Kinases genetics, Protein Processing, Post-Translational, Protein Serine-Threonine Kinases genetics, Signal Transduction, Bacterial Proteins metabolism, Protein Kinases metabolism, Protein Serine-Threonine Kinases metabolism, Tetratricopeptide Repeat
- Abstract
Signal transduction is essential for bacteria to adapt to changing environmental conditions. Among many forms of posttranslational modifications, reversible protein phosphorylation has evolved as a ubiquitous molecular mechanism of protein regulation in response to specific stimuli. The Ser/Thr protein kinase PknG modulates the fate of intracellular glutamate by controlling the phosphorylation status of the 2-oxoglutarate dehydrogenase regulator OdhI, a function that is conserved among diverse actinobacteria. PknG has a modular organization characterized by the presence of regulatory domains surrounding the catalytic domain. Here, we present an investigation using in vivo experiments, as well as biochemical and structural methods, of the molecular basis of the regulation of PknG from Corynebacterium glutamicum ( Cg PknG), in the light of previous knowledge available for the kinase from Mycobacterium tuberculosis ( Mtb PknG). We found that OdhI phosphorylation by Cg PknG is regulated by a conserved mechanism that depends on a C-terminal domain composed of tetratricopeptide repeats (TPRs) essential for metabolic homeostasis. Furthermore, we identified a conserved structural motif that physically connects the TPR domain to a β-hairpin within the flexible N-terminal region that is involved in docking interactions with OdhI. Based on our results and previous reports, we propose a model in which the TPR domain of PknG couples signal detection to the specific phosphorylation of OdhI. Overall, the available data indicate that conserved PknG domains in distant actinobacteria retain their roles in kinase regulation in response to nutrient availability. IMPORTANCE Bacteria control the metabolic processes by which they obtain nutrients and energy in order to adapt to the environment. Actinobacteria , one of the largest bacterial phyla of major importance for biotechnology, medicine, and agriculture, developed a unique control process that revolves around a key protein, the protein kinase PknG. Here, we use genetic, biochemical, and structural approaches to study PknG in a system that regulates glutamate production in Corynebacterium glutamicum, a species used for the industrial production of amino acids. The reported findings are conserved in related Actinobacteria , with broader significance for microorganisms that cause disease, as well as environmental species used industrially to produce amino acids and antibiotics every year.
- Published
- 2021
- Full Text
- View/download PDF
8. Epistatic interactions promote persistence of NS3-Q80K in HCV infection by compensating for protein folding instability.
- Author
-
Dultz G, Srikakulam SK, Konetschnik M, Shimakami T, Doncheva NT, Dietz J, Sarrazin C, Biondi RM, Zeuzem S, Tampé R, Kalinina OV, and Welsch C
- Subjects
- Amino Acid Substitution, Genes, Viral, Humans, Molecular Dynamics Simulation, Mutation, Polymorphism, Genetic, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins genetics, Epistasis, Genetic, Hepacivirus genetics, Hepatitis C virology
- Abstract
The Q80K polymorphism in the NS3-4A protease of the hepatitis C virus is associated with treatment failure of direct-acting antiviral agents. This polymorphism is highly prevalent in genotype 1a infections and stably transmitted between hosts. Here, we investigated the underlying molecular mechanisms of evolutionarily conserved coevolving amino acids in NS3-Q80K and revealed potential implications of epistatic interactions in immune escape and variants persistence. Using purified protein, we characterized the impact of epistatic amino acid substitutions on the physicochemical properties and peptide cleavage kinetics of the NS3-Q80K protease. We found that Q80K destabilized the protease protein fold (p < 0.0001). Although NS3-Q80K showed reduced peptide substrate turnover (p < 0.0002), replicative fitness in an H77S.3 cell culture model of infection was not significantly inferior to the WT virus. Epistatic substitutions at residues 91 and 174 in NS3-Q80K stabilized the protein fold (p < 0.0001) and leveraged the WT protease stability. However, changes in protease stability inversely correlated with enzymatic activity. In infectious cell culture, these secondary substitutions were not associated with a gain of replicative fitness in NS3-Q80K variants. Using molecular dynamics, we observed that the total number of residue contacts in NS3-Q80K mutants correlated with protein folding stability. Changes in the number of contacts reflected the compensatory effect on protein folding instability by epistatic substitutions. In summary, epistatic substitutions in NS3-Q80K contribute to viral fitness by mechanisms not directly related to RNA replication. By compensating for protein-folding instability, epistatic interactions likely protect NS3-Q80K variants from immune cell recognition., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
9. Crizotinib acts as ABL1 inhibitor combining ATP-binding with allosteric inhibition and is active against native BCR-ABL1 and its resistance and compound mutants BCR-ABL1 T315I and BCR-ABL1 T315I-E255K .
- Author
-
Mian AA, Haberbosch I, Khamaisie H, Agbarya A, Pietsch L, Eshel E, Najib D, Chiriches C, Ottmann OG, Hantschel O, Biondi RM, Ruthardt M, and Mahajna J
- Subjects
- Adenosine Triphosphate metabolism, Allosteric Regulation drug effects, Animals, Cell Line, Tumor, Drug Resistance, Neoplasm, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl metabolism, Humans, Jurkat Cells, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Mice, Mutation drug effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-abl metabolism, Antineoplastic Agents pharmacology, Crizotinib pharmacology, Fusion Proteins, bcr-abl antagonists & inhibitors, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Proto-Oncogene Proteins c-abl antagonists & inhibitors
- Abstract
Resistance remains the major clinical challenge for the therapy of Philadelphia chromosome-positive (Ph+) leukemia. With the exception of ponatinib, all approved tyrosine kinase inhibitors (TKIs) are unable to inhibit the common "gatekeeper" mutation T315I. Here we investigated the therapeutic potential of crizotinib, a TKI approved for targeting ALK and ROS1 in non-small cell lung cancer patients, which inhibited also the ABL1 kinase in cell-free systems, for the treatment of advanced and therapy-resistant Ph+ leukemia. By inhibiting the BCR-ABL1 kinase, crizotinib efficiently suppressed growth of Ph+ cells without affecting growth of Ph- cells. It was also active in Ph+ patient-derived long-term cultures (PD-LTCs) independently of the responsiveness/resistance to other TKIs. The efficacy of crizotinib was confirmed in vivo in syngeneic mouse models of BCR-ABL1- or BCR-ABL1
T315I -driven chronic myeloid leukemia-like disease and in BCR-ABL1-driven acute lymphoblastic leukemia (ALL). Although crizotinib binds to the ATP-binding site, it also allosterically affected the myristol binding pocket, the binding site of GNF2 and asciminib (former ABL001). Therefore, crizotinib has a seemingly unique double mechanism of action, on the ATP-binding site and on the myristoylation binding pocket. These findings strongly suggest the clinical evaluation of crizotinib for the treatment of advanced and therapy-resistant Ph+ leukemia., (© 2020. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
10. Demonstrating Ligandability of the LC3A and LC3B Adapter Interface.
- Author
-
Hartmann M, Huber J, Kramer JS, Heering J, Pietsch L, Stark H, Odadzic D, Bischoff I, Fürst R, Schröder M, Akutsu M, Chaikuad A, Dötsch V, Knapp S, Biondi RM, Rogov VV, and Proschak E
- Subjects
- 4-Hydroxycoumarins chemical synthesis, 4-Hydroxycoumarins metabolism, HEK293 Cells, Humans, Ligands, Molecular Structure, Novobiocin chemistry, Structure-Activity Relationship, 4-Hydroxycoumarins pharmacology, Autophagy drug effects, Microtubule-Associated Proteins metabolism, Protein Binding drug effects, Sequestosome-1 Protein metabolism
- Abstract
Autophagy is the common name for a number of lysosome-based degradation pathways of cytosolic cargos. The key components of autophagy are members of Atg8 family proteins involved in almost all steps of the process, from autophagosome formation to their selective fusion with lysosomes. In this study, we show that the homologous members of the human Atg8 family proteins, LC3A and LC3B, are druggable by a small molecule inhibitor novobiocin. Structure-activity relationship (SAR) studies of the 4-hydroxy coumarin core scaffold were performed, supported by a crystal structure of the LC3A dihydronovobiocin complex. The study reports the first nonpeptide inhibitors for these protein interaction targets and will lay the foundation for the development of more potent chemical probes for the Atg8 protein family which may also find applications for the development of autophagy-mediated degraders (AUTACs).
- Published
- 2021
- Full Text
- View/download PDF
11. Identification of Key Phospholipids That Bind and Activate Atypical PKCs.
- Author
-
Velnati S, Centonze S, Girivetto F, Capello D, Biondi RM, Bertoni A, Cantello R, Ragnoli B, Malerba M, Graziani A, and Baldanzi G
- Abstract
PKCζ and PKCι/λ form the atypical protein kinase C subgroup, characterised by a lack of regulation by calcium and the neutral lipid diacylglycerol. To better understand the regulation of these kinases, we systematically explored their interactions with various purified phospholipids using the lipid overlay assays, followed by kinase activity assays to evaluate the lipid effects on their enzymatic activity. We observed that both PKCζ and PKCι interact with phosphatidic acid and phosphatidylserine. Conversely, PKCι is unique in binding also to phosphatidylinositol-monophosphates (e.g., phosphatidylinositol 3-phosphate, 4-phosphate, and 5-phosphate). Moreover, we observed that phosphatidylinositol 4-phosphate specifically activates PKCι, while both isoforms are responsive to phosphatidic acid and phosphatidylserine. Overall, our results suggest that atypical Protein kinase C (PKC) localisation and activity are regulated by membrane lipids distinct from those involved in conventional PKCs and unveil a specific regulation of PKCι by phosphatidylinositol-monophosphates.
- Published
- 2021
- Full Text
- View/download PDF
12. Alternative AKT2 splicing produces protein lacking the hydrophobic motif regulatory region.
- Author
-
Plotz G, Lopez-Garcia LA, Brieger A, Zeuzem S, and Biondi RM
- Subjects
- Cell Line, Tumor, Cell Proliferation genetics, HEK293 Cells, Humans, Hydrophobic and Hydrophilic Interactions, Nucleotide Motifs genetics, Phosphatidylinositol 3-Kinases genetics, Phosphorylation genetics, Protein Isoforms genetics, Signal Transduction genetics, Alternative Splicing genetics, Proto-Oncogene Proteins c-akt genetics, Regulatory Sequences, Nucleic Acid genetics
- Abstract
Three AKT serine/threonine kinase isoforms (AKT1/AKT2/AKT3) mediate proliferation, metabolism, differentiation and anti-apoptotic signals. AKT isoforms are activated downstream of PI3-kinase and also by PI3-kinase independent mechanisms. Mutations in the lipid phosphatase PTEN and PI3-kinase that increase PIP3 levels increase AKT signaling in a large proportion of human cancers. AKT and other AGC kinases possess a regulatory mechanism that relies on a conserved hydrophobic motif (HM) C-terminal to the catalytic core. In AKT, the HM is contiguous to the serine 473 and two other newly discovered (serine 477 and tyrosine 479) regulatory phosphorylation sites. In AKT genes, this regulatory HM region is encoded in the final exon. We identified a splice variant of AKT2 (AKT2-13a), which contains an alternative final exon and lacks the HM regulatory site. We validated the presence of mRNA for this AKT2-13a splice variant in different tissues, and the presence of AKT2-13a protein in extracts from HEK293 cells. When overexpressed in HEK293 cells, AKT2-13a is phosphorylated at the activation loop and at the zipper/turn motif phosphorylation sites but has reduced specific activity. Analysis of the human transcriptome corresponding to other AGC kinases revealed that all three AKT isoforms express alternative transcripts lacking the HM regulatory motif, which was not the case for SGK1-3, S6K1-2, and classical, novel and atypical PKC isoforms. The transcripts of splice variants of Akt1-3 excluding the HM regulatory region could lead to expression of deregulated forms of AKT., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
13. Synergistic Allostery in Multiligand-Protein Interactions.
- Author
-
Ghode A, Gross LZF, Tee WV, Guarnera E, Berezovsky IN, Biondi RM, and Anand GS
- Subjects
- Allosteric Regulation, Allosteric Site, Binding Sites, Ligands, Peptides, Protein Kinases metabolism
- Abstract
Amide hydrogen-deuterium exchange mass spectrometry is powerful for describing combinatorial coupling effects of a cooperative ligand pair binding at noncontiguous sites: adenosine at the ATP-pocket and a docking peptide (PIFtide) at the PIF-pocket, on a model protein kinase PDK1. Binding of two ligands to PDK1 reveal multiple hotspots of synergistic allostery with cumulative effects greater than the sum of individual effects mediated by each ligand. We quantified this synergism and ranked these hotspots using a difference in deuteration-based approach, which showed that the strongest synergistic effects were observed at three of the critical catalytic loci of kinases: the αB-αC helices, and HRD-motif loop, and DFG-motif. Additionally, we observed weaker synergistic effects at a distal GHI-subdomain locus. Synergistic changes in deuterium exchange observed at a distal site but not at the intermediate sites of the large lobe of the kinase reveals allosteric propagation in proteins to operate through two modes. Direct electrostatic interactions between polar and charged amino acids that mediate targeted relay of allosteric signals, and diffused relay of allosteric signals through soft matter-like hydrophobic core amino acids. Furthermore, we provide evidence that the conserved β-3 strand lysine of protein kinases (Lys111 of PDK1) functions as an integrator node to coordinate allosteric coupling of the two ligand-binding sites. It maintains indirect interactions with the ATP-pocket and mediates a critical salt bridge with a glutamate (Glu130) of αC helix, which is conserved across all kinases. In summary, allosteric propagation in cooperative, dual-liganded enzyme targets is bidirectional and synergistic and offers a strategy for combinatorial drug development., (Copyright © 2020 Biophysical Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
14. Extended interaction networks with HCV protease NS3-4A substrates explain the lack of adaptive capability against protease inhibitors.
- Author
-
Dultz G, Shimakami T, Schneider M, Murai K, Yamane D, Marion A, Zeitler TM, Stross C, Grimm C, Richter RM, Bäumer K, Yi M, Biondi RM, Zeuzem S, Tampé R, Antes I, Lange CM, and Welsch C
- Subjects
- Amino Acid Substitution, Cell Line, Tumor, Humans, Mutation, Missense, Serine Proteases chemistry, Serine Proteases genetics, Serine Proteases metabolism, Viral Nonstructural Proteins antagonists & inhibitors, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins genetics, Viral Nonstructural Proteins metabolism, Virus Replication genetics, Adaptor Proteins, Signal Transducing chemistry, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Hepacivirus physiology, Molecular Dynamics Simulation, Protease Inhibitors pharmacology, Virus Replication drug effects
- Abstract
Inhibitors against the NS3-4A protease of hepatitis C virus (HCV) have proven to be useful drugs in the treatment of HCV infection. Although variants have been identified with mutations that confer resistance to these inhibitors, the mutations do not restore replicative fitness and no secondary mutations that rescue fitness have been found. To gain insight into the molecular mechanisms underlying the lack of fitness compensation, we screened known resistance mutations in infectious HCV cell culture with different genomic backgrounds. We observed that the Q41R mutation of NS3-4A efficiently rescues the replicative fitness in cell culture for virus variants containing mutations at NS3-Asp
168 To understand how the Q41R mutation rescues activity, we performed protease activity assays complemented by molecular dynamics simulations, which showed that protease-peptide interactions far outside the targeted peptide cleavage sites mediate substrate recognition by NS3-4A and support protease cleavage kinetics. These interactions shed new light on the mechanisms by which NS3-4A cleaves its substrates, viral polyproteins and a prime cellular antiviral adaptor protein, the mitochondrial antiviral signaling protein MAVS. Peptide binding is mediated by an extended hydrogen-bond network in NS3-4A that was effectively optimized for protease-MAVS binding in Asp168 variants with rescued replicative fitness from NS3-Q41R. In the protease harboring NS3-Q41R, the N-terminal cleavage products of MAVS retained high affinity to the active site, rendering the protease susceptible for potential product inhibition. Our findings reveal delicately balanced protease-peptide interactions in viral replication and immune escape that likely restrict the protease adaptive capability and narrow the virus evolutionary space., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Dultz et al.)- Published
- 2020
- Full Text
- View/download PDF
15. ACE2, the Receptor that Enables Infection by SARS-CoV-2: Biochemistry, Structure, Allostery and Evaluation of the Potential Development of ACE2 Modulators.
- Author
-
Gross LZF, Sacerdoti M, Piiper A, Zeuzem S, Leroux AE, and Biondi RM
- Subjects
- Allosteric Regulation, Angiotensin-Converting Enzyme 2, Angiotensin-Converting Enzyme Inhibitors chemistry, Catalytic Domain, Humans, Peptidyl-Dipeptidase A chemistry, Protein Binding, Protein Domains, Receptors, Virus antagonists & inhibitors, Receptors, Virus chemistry, SARS-CoV-2, Spike Glycoprotein, Coronavirus chemistry, Angiotensin-Converting Enzyme Inhibitors metabolism, Betacoronavirus chemistry, Peptidyl-Dipeptidase A metabolism, Receptors, Virus metabolism, Spike Glycoprotein, Coronavirus metabolism
- Abstract
Angiotensin converting enzyme 2 (ACE2) is the human receptor that interacts with the spike protein of coronaviruses, including the one that produced the 2020 coronavirus pandemic (COVID-19). Thus, ACE2 is a potential target for drugs that disrupt the interaction of human cells with SARS-CoV-2 to abolish infection. There is also interest in drugs that inhibit or activate ACE2, that is, for cardiovascular disorders or colitis. Compounds binding at alternative sites could allosterically affect the interaction with the spike protein. Herein, we review biochemical, chemical biology, and structural information on ACE2, including the recent cryoEM structures of full-length ACE2. We conclude that ACE2 is very dynamic and that allosteric drugs could be developed to target ACE2. At the time of the 2020 pandemic, we suggest that available ACE2 inhibitors or activators in advanced development should be tested for their ability to allosterically displace the interaction between ACE2 and the spike protein., (© 2020 Wiley-VCH GmbH.)
- Published
- 2020
- Full Text
- View/download PDF
16. Renaissance of Allostery to Disrupt Protein Kinase Interactions.
- Author
-
Leroux AE and Biondi RM
- Subjects
- Adenosine Triphosphate chemistry, Allosteric Regulation, Humans, Protein Binding, Protein Kinases chemistry, Adenosine Triphosphate metabolism, Protein Kinases metabolism
- Abstract
Protein-protein interactions often regulate the activity of protein kinases by allosterically modulating the conformation of the ATP-binding site. Bidirectional allostery implies that reverse modulation (i.e., from the ATP-binding site to the interaction and regulatory sites) must also be possible. Here, we review both the allosteric regulation of protein kinases and recent work describing how compounds binding at the ATP-binding site can promote or inhibit protein kinase interactions at regulatory sites via the reverse mechanism. Notably, the pharmaceutical industry has been developing compounds that bind to the ATP-binding site of protein kinases and potently disrupt protein-protein interactions between target protein kinases and their regulatory interacting partners. Learning to modulate allosteric processes will facilitate the development of protein-protein interaction modulators., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
17. Allosteric Regulation of Protein Kinases Downstream of PI3-Kinase Signalling.
- Author
-
Leroux AE, Gross LZF, Sacerdoti M, and Biondi RM
- Subjects
- Allosteric Regulation, Allosteric Site, Humans, Protein Binding drug effects, Protein Kinase Inhibitors pharmacology, Drug Development, Phosphatidylinositol 3-Kinases metabolism, Protein Kinases metabolism, Signal Transduction drug effects
- Abstract
Allostery is a basic principle that enables proteins to process and transmit cellular information. Protein kinases evolved allosteric mechanisms to transduce cellular signals to downstream signalling components or effector molecules. Protein kinases catalyse the transfer of the terminal phosphate from ATP to protein substrates upon specific stimuli. Protein kinases are targets for the development of small molecule inhibitors for the treatment of human diseases. Drug development has focussed on ATP-binding site, while there is increase interest in the development of drugs targeting alternative sites, i.e. allosteric sites. Here, we review the mechanism of regulation of protein kinases, which often involve the allosteric modulation of the ATP-binding site, enhancing or inhibiting activity. We exemplify the molecular mechanism of allostery in protein kinases downstream of PI3-kinase signalling with a focus on phosphoinositide-dependent protein kinase 1 (PDK1), a model kinase where small compounds can allosterically modulate the conformation of the kinase bidirectionally.
- Published
- 2019
- Full Text
- View/download PDF
18. Predicting protein targets for drug-like compounds using transcriptomics.
- Author
-
Pabon NA, Xia Y, Estabrooks SK, Ye Z, Herbrand AK, Süß E, Biondi RM, Assimon VA, Gestwicki JE, Brodsky JL, Camacho CJ, and Bar-Joseph Z
- Subjects
- Cell Line, Computational Biology, Computer Simulation, Databases, Nucleic Acid statistics & numerical data, Drug Discovery statistics & numerical data, Drug Evaluation, Preclinical methods, Drug Evaluation, Preclinical statistics & numerical data, Gene Expression Profiling statistics & numerical data, Gene Knockdown Techniques, Gene Ontology, Gene Regulatory Networks drug effects, Humans, Models, Molecular, Molecular Docking Simulation, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacology, Proteins genetics, Ubiquitin-Protein Ligases antagonists & inhibitors, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases genetics, Wortmannin chemistry, Wortmannin pharmacology, ras Proteins antagonists & inhibitors, ras Proteins chemistry, ras Proteins genetics, Drug Discovery methods, Gene Expression Profiling methods, Proteins chemistry, Proteins drug effects
- Abstract
An expanded chemical space is essential for improved identification of small molecules for emerging therapeutic targets. However, the identification of targets for novel compounds is biased towards the synthesis of known scaffolds that bind familiar protein families, limiting the exploration of chemical space. To change this paradigm, we validated a new pipeline that identifies small molecule-protein interactions and works even for compounds lacking similarity to known drugs. Based on differential mRNA profiles in multiple cell types exposed to drugs and in which gene knockdowns (KD) were conducted, we showed that drugs induce gene regulatory networks that correlate with those produced after silencing protein-coding genes. Next, we applied supervised machine learning to exploit drug-KD signature correlations and enriched our predictions using an orthogonal structure-based screen. As a proof-of-principle for this regimen, top-10/top-100 target prediction accuracies of 26% and 41%, respectively, were achieved on a validation of set 152 FDA-approved drugs and 3104 potential targets. We then predicted targets for 1680 compounds and validated chemical interactors with four targets that have proven difficult to chemically modulate, including non-covalent inhibitors of HRAS and KRAS. Importantly, drug-target interactions manifest as gene expression correlations between drug treatment and both target gene KD and KD of genes that act up- or down-stream of the target, even for relatively weak binders. These correlations provide new insights on the cellular response of disrupting protein interactions and highlight the complex genetic phenotypes of drug treatment. With further refinement, our pipeline may accelerate the identification and development of novel chemical classes by screening compound-target interactions., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
19. DNA mismatch repair activity of MutLα is regulated by CK2-dependent phosphorylation of MLH1 (S477).
- Author
-
Weßbecher IM, Hinrichsen I, Funke S, Oellerich T, Plotz G, Zeuzem S, Grus FH, Biondi RM, and Brieger A
- Subjects
- Animals, Cell Cycle, Cell Line, Tumor, DNA Mismatch Repair, Gene Expression Regulation, Neoplastic, HEK293 Cells, Humans, Mass Spectrometry, Mismatch Repair Endonuclease PMS2 chemistry, Mismatch Repair Endonuclease PMS2 metabolism, Models, Molecular, MutL Proteins chemistry, Phosphorylation, Protein Processing, Post-Translational, Serine metabolism, Sf9 Cells, Casein Kinase II metabolism, MutL Protein Homolog 1 chemistry, MutL Protein Homolog 1 metabolism, MutL Proteins metabolism
- Abstract
MutLα, a heterodimer consisting of MLH1 and PMS2, is a key player of DNA mismatch repair (MMR), yet little is known about its regulation. In this study, we used mass spectrometry to identify phosphorylated residues within MLH1 and PMS2. The most frequently detected phosphorylated amino acid was serine 477 of MLH1. Pharmacological treatment indicates that Casein kinase II (CK2) could be responsible for the phosphorylation of MLH1 at serine 477 in vivo. In vitro kinase assay verified MLH1 as a substrate of CK2. Most importantly, using in vitro MMR assay we could demonstrate that p-MLH1
S477 lost MMR activity. Moreover, we found that levels of p-MLH1S477 varied during the cell cycle. In summary, we identified that phosphorylation of MLH1 by CK2 at amino acid position 477 can switch off MMR activity in vitro. Since CK2 is overexpressed in many tumors and is able to inactivate MMR, the new mechanism here described could have an important impact on tumors overactive in CK2., (© 2018 Wiley Periodicals, Inc.)- Published
- 2018
- Full Text
- View/download PDF
20. Modulation of the Allosteric Communication between the Polo-Box Domain and the Catalytic Domain in Plk1 by Small Compounds.
- Author
-
Raab M, Sanhaji M, Pietsch L, Béquignon I, Herbrand AK, Süß E, Gande SL, Caspar B, Kudlinzki D, Saxena K, Sreeramulu S, Schwalbe H, Strebhardt K, and Biondi RM
- Subjects
- Allosteric Regulation drug effects, Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Apoptosis drug effects, Catalytic Domain, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Proliferation drug effects, Centrosome metabolism, Enzyme Activators pharmacology, G2 Phase Cell Cycle Checkpoints drug effects, HeLa Cells, Humans, Kinetochores metabolism, Oligopeptides chemistry, Phosphopeptides chemistry, Phosphopeptides metabolism, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins metabolism, Small Molecule Libraries pharmacology, Spodoptera chemistry, Polo-Like Kinase 1, Cell Cycle Proteins agonists, Cell Cycle Proteins antagonists & inhibitors, Enzyme Activators chemistry, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins agonists, Proto-Oncogene Proteins antagonists & inhibitors, Small Molecule Libraries chemistry
- Abstract
The Polo-like kinases (Plks) are an evolutionary conserved family of Ser/Thr protein kinases that possess, in addition to the classical kinase domain at the N-terminus, a C-terminal polo-box domain (PBD) that binds to phosphorylated proteins and modulates the kinase activity and its localization. Plk1, which regulates the formation of the mitotic spindle, has emerged as a validated drug target for the treatment of cancer, because it is required for numerous types of cancer cells but not for the cell division in noncancer cells. Here, we employed chemical biology methods to investigate the allosteric communication between the PBD and the catalytic domain of Plk1. We identified small compounds that bind to the catalytic domain and inhibit or enhance the interaction of Plk1 with the phosphorylated peptide PoloBoxtide in vitro. In cells, two new allosteric Plk1 inhibitors affected the proliferation of cancer cells in culture and the cell cycle but had distinct phenotypic effects on spindle formation. Both compounds inhibited Plk1 signaling, indicating that they specifically act on Plk1 in cultured cells.
- Published
- 2018
- Full Text
- View/download PDF
21. AGC kinases, mechanisms of regulation and innovative drug development.
- Author
-
Leroux AE, Schulze JO, and Biondi RM
- Subjects
- Drug Discovery, Humans, Phosphorylation, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, rho-Associated Kinases metabolism, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases physiology
- Abstract
The group of AGC kinases consists of 63 evolutionarily related serine/threonine protein kinases comprising PDK1, PKB/Akt, SGK, PKC, PRK/PKN, MSK, RSK, S6K, PKA, PKG, DMPK, MRCK, ROCK, NDR, LATS, CRIK, MAST, GRK, Sgk494, and YANK, while two other families, Aurora and PLK, are the most closely related to the group. Eight of these families are physiologically activated downstream of growth factor signalling, while other AGC kinases are downstream effectors of a wide range of signals. The different AGC kinase families share aspects of their mechanisms of inhibition and activation. In the present review, we update the knowledge of the mechanisms of regulation of different AGC kinases. The conformation of the catalytic domain of many AGC kinases is regulated allosterically through the modulation of the conformation of a regulatory site on the small lobe of the kinase domain, the PIF-pocket. The PIF-pocket acts like an ON-OFF switch in AGC kinases with different modes of regulation, i.e. PDK1, PKB/Akt, LATS and Aurora kinases. In this review, we make emphasis on how the knowledge of the molecular mechanisms of regulation can guide the discovery and development of small allosteric modulators. Molecular probes stabilizing the PIF-pocket in the active conformation are activators, while compounds stabilizing the disrupted site are allosteric inhibitors. One challenge for the rational development of allosteric modulators is the lack of complete structural information of the inhibited forms of full-length AGC kinases. On the other hand, we suggest that the available information derived from molecular biology and biochemical studies can already guide screening strategies for the identification of innovative mode of action molecular probes and the development of selective allosteric drugs for the treatment of human diseases., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
22. Activation of Adenylyl Cyclase Causes Stimulation of Adenosine Receptors.
- Author
-
Pleli T, Mondorf A, Ferreiros N, Thomas D, Dvorak K, Biondi RM, Heringdorf DMZ, Zeuzem S, Geisslinger G, Zimmermann H, Waidmann O, and Piiper A
- Subjects
- Animals, Cyclic AMP metabolism, Enzyme Activation, GTP-Binding Protein alpha Subunits, Gs metabolism, PC12 Cells, Rats, Signal Transduction, Adenylyl Cyclases metabolism, Receptor, Adenosine A2A metabolism, Receptor, Adenosine A2B metabolism
- Abstract
Background/aims: Signaling of Gs protein-coupled receptors (GsPCRs) is accomplished by stimulation of adenylyl cyclase, causing an increase of the intracellular cAMP concentration, activation of the intracellular cAMP effectors protein kinase A (PKA) and Epac, and an efflux of cAMP, the function of which is still unclear., Methods: Activation of adenylyl cyclase by GsPCR agonists or cholera toxin was monitored by measurement of the intracellular cAMP concentration by ELISA, anti-phospho-PKA substrate motif phosphorylation by immunoblotting, and an Epac-FRET assay in the presence and absence of adenosine receptor antagonists or ecto-nucleotide phosphodiesterase/pyrophosphatase2 (eNPP2) inhibitors. The production of AMP from cAMP by recombinant eNPP2 was measured by HPLC. Extracellular adenosine was determined by LC-MS/MS, extracellular ATP by luciferase and LC-MS/MS. The expression of eNPP isoenzymes 1-3 was examined by RT-PCR. The expression of multidrug resistance protein 4 was suppressed by siRNA., Results: Here we show that the activation of GsPCRs and the GsPCRs-independent activation of Gs proteins and adenylyl cyclase by cholera toxin induce stimulation of cell surface adenosine receptors (A2A or A2B adenosine receptors). In PC12 cells stimulation of adenylyl cyclase by GsPCR or cholera toxin caused activation of A2A adenosine receptors by an autocrine signaling pathway involving cAMP efflux through multidrug resistance protein 4 and hydrolysis of released cAMP to AMP by eNPP2. In contrast, in PC3 cells cholera toxin- and GsPCR-induced stimulation of adenylyl cyclase resulted in the activation of A2B adenosine receptors., Conclusion: Our findings show that stimulation of adenylyl cyclase causes a remarkable activation of cell surface adenosine receptors., (© 2018 The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2018
- Full Text
- View/download PDF
23. Camptothecin and its analog SN-38, the active metabolite of irinotecan, inhibit binding of the transcriptional regulator and oncoprotein FUBP1 to its DNA target sequence FUSE.
- Author
-
Khageh Hosseini S, Kolterer S, Steiner M, von Manstein V, Gerlach K, Trojan J, Waidmann O, Zeuzem S, Schulze JO, Hahn S, Steinhilber D, Gatterdam V, Tampé R, Biondi RM, Proschak E, and Zörnig M
- Subjects
- Antineoplastic Agents, Phytogenic metabolism, Camptothecin metabolism, Cell Line, Tumor, DNA chemistry, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, HEK293 Cells, Humans, Irinotecan, Protein Binding, RNA-Binding Proteins, Antineoplastic Agents, Phytogenic pharmacology, Camptothecin analogs & derivatives, Camptothecin pharmacology, DNA genetics, DNA Helicases metabolism, DNA-Binding Proteins metabolism
- Abstract
The transcriptional regulator FUSE Binding Protein 1 (FUBP1) is overexpressed in more than 80% of all human hepatocellular carcinomas (HCCs) and other solid tumor entities including prostate and colorectal carcinoma. FUBP1 expression is required for HCC tumor cell expansion, and it functions as an important pro-proliferative and anti-apoptotic oncoprotein that binds to the single-stranded DNA sequence FUSE to regulate the transcription of a variety of target genes. In this study, we screened an FDA-approved drug library and discovered that the Topoisomerase I (TOP1) inhibitor camptothecin (CPT) and its derivative 7-ethyl-10-hydroxycamptothecin (SN-38), the active irinotecan metabolite that is used in the clinics in combination with other chemotherapeutics to treat carcinoma, inhibit FUBP1 activity. Both molecules prevent in vitro the binding of FUBP1 to its single-stranded target DNA FUSE, and they induce deregulation of FUBP1 target genes in HCC cells. Our results suggest the interference with the FUBP1/FUSE interaction as a further molecular mechanism that, in addition to the inactivation of TOP1, may contribute to the therapeutic potential of CPT/SN-38. Targeting of FUBP1 in HCC therapy with SN-38/irinotecan could be a particularly interesting option because of the high FUBP1 levels in HCC cells and their dependency on FUBP1 expression., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
24. Phosphorylation-dependent signaling controls degradation of DNA mismatch repair protein PMS2.
- Author
-
Hinrichsen I, Weßbecher IM, Huhn M, Passmann S, Zeuzem S, Plotz G, Biondi RM, and Brieger A
- Subjects
- Blotting, Western, Enzyme Inhibitors pharmacology, HEK293 Cells, Humans, Marine Toxins, Mismatch Repair Endonuclease PMS2 genetics, MutL Protein Homolog 1 genetics, MutL Proteins genetics, Mutation, Niacinamide analogs & derivatives, Niacinamide pharmacology, Oxazoles pharmacology, Phenylurea Compounds pharmacology, Phosphorylation, Proteolysis drug effects, Serine genetics, Serine metabolism, Sorafenib, Threonine genetics, Threonine metabolism, Mismatch Repair Endonuclease PMS2 metabolism, MutL Protein Homolog 1 metabolism, MutL Proteins metabolism, Signal Transduction
- Abstract
MutLα, a heterodimer consisting of MLH1 and PMS2, plays an important role in DNA mismatch repair and has been shown to be additionally involved in several other important cellular mechanisms. Previous work indicated that AKT could modulate PMS2 stability by phosphorylation. Still, the mechanisms of regulation of MutLα remain unclear. The stability of MutLα subunits was investigated by transiently overexpression of wild type and mutant forms of MLH1 and PMS2 using immunoblotting for measuring the protein levels after treatment. We found that treatment with the cell-permeable serine/threonine phosphatase inhibitor, Calyculin, leads to degradation of PMS2 when MLH1 or its C-terminal domain is missing or if amino acids of MLH1 essential for PMS2 interaction are mutated. In addition, we discovered that the C-terminal tail of PMS2 is relevant for this Calyculin-dependent degradation. A direct involvement of AKT, which was previously described to be responsible for PMS2 degradation, could not be detected. The multi-kinase inhibitor Sorafenib, in contrast, was able to avoid the degradation of PMS2 which postulates that cellular phosphorylation is involved in this process. Together, we show that pharmacologically induced phosphorylation by Calyculin can induce the selective proteasome-dependent degradation of PMS2 but not of MLH1 and that the PMS2 degradation could be blocked by Sorafenib treatment. Curiously, the C-terminal Lynch Syndrome-variants MLH1
L749P and MLH1Y750X make PMS2 prone to Calyculin induced degradation. Therefore, we conclude that the specific degradation of PMS2 may represent a new mechanism to regulate MutLα., (© 2017 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
25. An Allosteric Inhibitor Scaffold Targeting the PIF-Pocket of Atypical Protein Kinase C Isoforms.
- Author
-
Arencibia JM, Fröhner W, Krupa M, Pastor-Flores D, Merker P, Oellerich T, Neimanis S, Schmithals C, Köberle V, Süß E, Zeuzem S, Stark H, Piiper A, Odadzic D, Schulze JO, and Biondi RM
- Subjects
- Allosteric Regulation, Animals, Cell Line, Tumor, Female, Heterografts, Humans, Lung Neoplasms enzymology, Lung Neoplasms pathology, Mice, Mice, Nude, Isoenzymes metabolism, Protein Kinase C metabolism
- Abstract
There is a current and pressing need for improved cancer therapies. The use of small molecule kinase inhibitors and their application in combinatorial regimens represent an approach to personalized targeted cancer therapy. A number of AGC kinases, including atypical Protein Kinase C enzymes (PKCs), are validated drug targets for cancer treatment. Most drug development programs for protein kinases focus on the development of drugs that bind at the ATP-binding site. Alternatively, allosteric drugs have great potential for the development of future innovative drugs. However, the rational development of allosteric drugs poses important challenges because the compounds not only must bind to a given site but also must stabilize forms of the protein with a desired effect at a distant site. Here we describe the development of a new class of compounds targeting a regulatory site (PIF-pocket) present in the kinase domain and provide biochemical and crystallographic data showing that these compounds allosterically inhibit the activity of atypical PKCs. PS432, a representative compound, decreased the rate of proliferation of non-small cell lung cancer cells more potently than aurothiomalate, an atypical PKCι inhibitor currently under evaluation in clinical trials, and significantly reduced tumor growth without side effects in a mouse xenograft model. The druglike chemical class provides ample possibilities for the synthesis of derivative compounds, with the potential to allosterically modulate the activity of atypical PKCs and other kinases.
- Published
- 2017
- Full Text
- View/download PDF
26. Pyrazolo[1,5a]pyrimidines as a new class of FUSE binding protein 1 (FUBP1) inhibitors.
- Author
-
Hauck S, Hiesinger K, Khageh Hosseini S, Achenbach J, Biondi RM, Proschak E, Zörnig M, and Odadzic D
- Subjects
- Cell Death drug effects, Dose-Response Relationship, Drug, Humans, Molecular Structure, Pyrazoles chemical synthesis, Pyrazoles chemistry, Pyrimidines chemical synthesis, Pyrimidines chemistry, RNA-Binding Proteins, Structure-Activity Relationship, Tumor Cells, Cultured, DNA Helicases antagonists & inhibitors, DNA-Binding Proteins antagonists & inhibitors, Pyrazoles pharmacology, Pyrimidines pharmacology
- Abstract
The transcriptional regulator FUSE binding protein 1 (FUBP1) is aberrantly upregulated in various malignancies, fulfilling its oncogenic role by the deregulation of critical genes involved in cell cycle control and apoptosis regulation. Thus, the pharmaceutical inhibition of this protein would represent an encouraging novel targeted chemotherapy. Here, we demonstrate the identification and initial optimization of a pyrazolo[1,5a]pyrimidine-based FUBP1 inhibitor derived from medium throughput screening, which interferes with the binding of FUBP1 to its single stranded target DNA FUSE. We were able to generate a new class of FUBP1 interfering molecules with in vitro and biological activity. In biophysical assays, we could show that our best inhibitor, compound 6, potently inhibits the binding of FUBP1 to the FUSE sequence with an IC
50 value of 11.0μM. Furthermore, hepatocellular carcinoma cells exhibited sensitivity towards the treatment with compound 6, resulting in reduced cell expansion and induction of cell death. Finally, we provide insights into the corresponding SAR landscape, leading to a prospective enhancement in potency and cellular efficacy., (Copyright © 2016. Published by Elsevier Ltd.)- Published
- 2016
- Full Text
- View/download PDF
27. Bidirectional Allosteric Communication between the ATP-Binding Site and the Regulatory PIF Pocket in PDK1 Protein Kinase.
- Author
-
Schulze JO, Saladino G, Busschots K, Neimanis S, Süß E, Odadzic D, Zeuzem S, Hindie V, Herbrand AK, Lisa MN, Alzari PM, Gervasio FL, and Biondi RM
- Subjects
- Allosteric Site drug effects, Aurora Kinases antagonists & inhibitors, Aurora Kinases chemistry, Aurora Kinases metabolism, Binding Sites drug effects, HEK293 Cells, Humans, Indazoles chemistry, Molecular Docking Simulation, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism, Pyrimidines chemistry, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Adenosine Triphosphate metabolism, Allosteric Regulation drug effects, Indazoles pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Pyrimidines pharmacology
- Abstract
Allostery is a phenomenon observed in many proteins where binding of a macromolecular partner or a small-molecule ligand at one location leads to specific perturbations at a site not in direct contact with the region where the binding occurs. The list of proteins under allosteric regulation includes AGC protein kinases. AGC kinases have a conserved allosteric site, the phosphoinositide-dependent protein kinase 1 (PDK1)-interacting fragment (PIF) pocket, which regulates protein ATP-binding, activity, and interaction with substrates. In this study, we identify small molecules that bind to the ATP-binding site and affect the PIF pocket of AGC kinase family members, PDK1 and Aurora kinase. We describe the mechanistic details and show that although PDK1 and Aurora kinase inhibitors bind to the conserved ATP-binding site, they differentially modulate physiological interactions at the PIF-pocket site. Our work outlines a strategy for developing bidirectional small-molecule allosteric modulators of protein kinases and other signaling proteins., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
28. Lipid regulators of Pkh2 in Candida albicans, the protein kinase ortholog of mammalian PDK1.
- Author
-
Pastor-Flores D, Schulze JO, Bahí A, Süß E, Casamayor A, and Biondi RM
- Subjects
- 3-Phosphoinositide-Dependent Protein Kinases genetics, Amino Acid Sequence, Binding Sites, Candida albicans genetics, Computational Biology, HEK293 Cells, Humans, Models, Molecular, Molecular Sequence Data, Phosphatidic Acids metabolism, Phosphatidylinositol 4,5-Diphosphate metabolism, Phosphatidylinositol Phosphates metabolism, Phosphatidylserines metabolism, Protein Binding, Protein Structure, Tertiary, Sphingosine analogs & derivatives, Sphingosine metabolism, Structure-Activity Relationship, Sulfoglycosphingolipids metabolism, Transfection, 3-Phosphoinositide-Dependent Protein Kinases metabolism, Candida albicans enzymology, Fungal Proteins metabolism, Lipid Metabolism
- Abstract
Pkh is the yeast ortholog of the mammalian 3-phosphoinositide-dependent protein kinase 1 (PDK1). Pkh phosphorylates the activation loop of Ypks, Tpks, Sch9 and also phosphorylates the eisosome components Lsp1 and Pil1, which play fundamental roles upstream of diverse signaling pathways, including the cell wall integrity and sphingosine/long-chain base (LCB) signaling pathways. In S. cerevisiae, two isoforms, ScPkh1 and ScPkh2, are required for cell viability, while only one ortholog exists in C. albicans, CaPkh2. In spite of the extensive information gathered on the role of Pkh in the LCB signaling, the yeast Pkh kinases are not known to bind lipids and previous studies did not identify PH domains in Pkh sequences. We now describe that the C-terminal region of CaPkh2 is required for its intrinsic kinase activity. In addition, we found that the C-terminal region of CaPkh2 enables its interaction with structural and signaling lipids. Our results further show that phosphatidylserine, phosphatidic acid, phosphatidylinositol (3,4 and 4,5)-biphosphates, and phosphatidylinositol (3,4,5)-trisphosphate inhibit Pkh activity, whereas sulfatide binds with high affinity but does not affect the intrinsic activity of CaPkh2. Interestingly, we identified that its human ortholog PDK1 also binds to sulfatide. We propose a mechanism by which lipids and dihydrosphingosine regulate CaPkh2 kinase activity by modulating the interaction of the C-terminal region with the kinase domain, while sulfatide-like lipids support localization CaPkh2 mediated by a C-terminal PH domain, without affecting kinase intrinsic activity., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
29. Discovery of a Potent Allosteric Kinase Modulator by Combining Computational and Synthetic Methods.
- Author
-
Kroon E, Schulze JO, Süß E, Camacho CJ, Biondi RM, and Dömling A
- Subjects
- Allosteric Regulation drug effects, Humans, Models, Molecular, Molecular Structure, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases metabolism, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Software, Drug Discovery methods, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
The rational design of allosteric kinase modulators is challenging but rewarding. The protein kinase PDK1, which lies at the center of the growth-factor signaling pathway, possesses an allosteric regulatory site previously validated both in vitro and in cells. ANCHOR.QUERY software was used to discover a potent allosteric PDK1 kinase modulator. Using a recently published PDK1 compound as a template, several new scaffolds that bind to the allosteric target site were generated and one example was validated. The inhibitor can be synthesized in one step by multicomponent reaction (MCR) chemistry when using the ANCHOR.QUERY approach. Our results are significant because the outlined approach allows rapid and efficient scaffold hopping from known molecules into new easily accessible and biologically active ones. Based on increasing interest in allosteric-site drug discovery, we foresee many potential applications for this approach., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
30. Depletion of yeast PDK1 orthologs triggers a stress-like transcriptional response.
- Author
-
Pastor-Flores D, Ferrer-Dalmau J, Bahí A, Boleda M, Biondi RM, and Casamayor A
- Subjects
- 3-Phosphoinositide-Dependent Protein Kinases metabolism, Cluster Analysis, Fermentation, Gene Expression Profiling, Genes, Lethal, Glycogen metabolism, Heat-Shock Response genetics, Ions, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Transcription Factors genetics, Transcription Factors metabolism, 3-Phosphoinositide-Dependent Protein Kinases genetics, Gene Expression Regulation, Fungal, Saccharomyces cerevisiae genetics, Stress, Physiological genetics, Transcription, Genetic
- Abstract
Background: Pkh proteins are the PDK1 orthologs in S. cerevisiae. They have redundant and essential activity and are responsible for the phosphorylation of several members of the AGC family of protein kinases. Pkh proteins have been involved in several cellular functions, including cell wall integrity and endocytosis. However the global expression changes caused by their depletion are still unknown., Results: A doxycycline-repressible tetO7 promoter driving the expression of PKH2 in cells carrying deletions of the PKH1 and PKH3 genes allowed us to progressively deplete cells from Pkh proteins when treated with doxycycline. Global gene expression analysis indicate that depletion of Pkh results in the up-regulation of genes involved in the accumulation of glycogen and also of those related to stress responses. Moreover, genes involved in the ion transport were quickly down-regulated when the levels of Pkh decreased. The reduction in the mRNA levels required for protein translation, however, was only observed after longer doxycycline treatment (24 h). We uncovered that Pkh is important for the proper transcriptional response to heat shock, and is mostly required for the effects driven by the transcription factors Hsf1 and Msn2/Msn4, but is not required for down-regulation of the mRNA coding for ribosomal proteins., Conclusions: By using the tetO7 promoter we elucidated for the first time the transcriptomic changes directly or indirectly caused by progressive depletion of Pkh. Furthermore, this system enabled the characterization of the transcriptional response triggered by heat shock in wild-type and Pkh-depleted cells, showing that about 40 % of the observed expression changes were, to some degree, dependent on Pkh.
- Published
- 2015
- Full Text
- View/download PDF
31. Molecular Basis of the Activity and the Regulation of the Eukaryotic-like S/T Protein Kinase PknG from Mycobacterium tuberculosis.
- Author
-
Lisa MN, Gil M, André-Leroux G, Barilone N, Durán R, Biondi RM, and Alzari PM
- Subjects
- Cloning, Molecular, Crystallization, Mutagenesis, Phosphorylation, Protein Structure, Tertiary, Rubredoxins chemistry, Rubredoxins metabolism, Substrate Specificity, Bacterial Proteins chemistry, Gene Expression Regulation, Enzymologic genetics, Models, Molecular, Mycobacterium tuberculosis enzymology, Protein Serine-Threonine Kinases chemistry
- Abstract
Tuberculosis remains one of the world's deadliest human diseases, with a high prevalence of antibiotic-resistant Mycobacterium tuberculosis (Mtb) strains. A molecular understanding of processes underlying regulation and adaptation of bacterial physiology may provide novel avenues for the development of antibiotics with unconventional modes of action. Here, we focus on the multidomain S/T protein kinase PknG, a soluble enzyme that controls central metabolism in Actinobacteria and has been linked to Mtb infectivity. Our biochemical and structural studies reveal how different motifs and domains flanking the catalytic core regulate substrate selectivity without significantly affecting the intrinsic kinase activity, whereas a rubredoxin-like domain is shown to downregulate catalysis through specific intramolecular interactions that modulate access to a profound substrate-binding site. Our findings provide the basis for the selective and specific inhibition of PknG, and open new questions about regulation of related bacterial and eukaryotic protein kinases., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. Molecular mechanism of regulation of the atypical protein kinase C by N-terminal domains and an allosteric small compound.
- Author
-
Zhang H, Neimanis S, Lopez-Garcia LA, Arencibia JM, Amon S, Stroba A, Zeuzem S, Proschak E, Stark H, Bauer AF, Busschots K, Jørgensen TJ, Engel M, Schulze JO, and Biondi RM
- Subjects
- Allosteric Regulation drug effects, Biphenyl Compounds chemistry, Cinnamates chemistry, Humans, Models, Molecular, Molecular Structure, Protein Kinase C antagonists & inhibitors, Protein Structure, Tertiary drug effects, Structure-Activity Relationship, Biphenyl Compounds pharmacology, Cinnamates pharmacology, Protein Kinase C chemistry, Protein Kinase C metabolism, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacology
- Abstract
Protein kinases play important regulatory roles in cells and organisms. Therefore, they are subject to specific and tight mechanisms of regulation that ultimately converge on the catalytic domain and allow the kinases to be activated or inhibited only upon the appropriate stimuli. AGC protein kinases have a pocket in the catalytic domain, the PDK1-interacting fragment (PIF)-pocket, which is a key mediator of the activation. We show here that helix αC within the PIF-pocket of atypical protein kinase C (aPKC) is the target of the interaction with its inhibitory N-terminal domains. We also provide structural evidence that the small compound PS315 is an allosteric inhibitor that binds to the PIF-pocket of aPKC. PS315 exploits the physiological dynamics of helix αC for its binding and allosteric inhibition. The results will support research on allosteric mechanisms and selective drug development efforts against PKC isoforms., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
33. Characterization of pomiferin triacetate as a novel mTOR and translation inhibitor.
- Author
-
Bajer MM, Kunze MM, Blees JS, Bokesch HR, Chen H, Brauss TF, Dong Z, Gustafson KR, Biondi RM, Henrich CJ, McMahon JB, Colburn NH, Schmid T, and Brüne B
- Subjects
- Apoptosis Regulatory Proteins metabolism, HEK293 Cells, Humans, MCF-7 Cells, Mechanistic Target of Rapamycin Complex 1, Mechanistic Target of Rapamycin Complex 2, Molecular Docking Simulation, Multiprotein Complexes metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, RNA-Binding Proteins metabolism, Ribosomal Protein S6 Kinases, 70-kDa metabolism, TOR Serine-Threonine Kinases metabolism, Isoflavones pharmacology, Protein Biosynthesis drug effects, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Deregulation of the phosphatidylinositol 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR)-70kDa ribosomal protein S6 kinase 1 (p70(S6K)) pathway is commonly observed in many tumors. This pathway controls proliferation, survival, and translation, and its overactivation is associated with poor prognosis for tumor-associated survival. Current efforts focus on the development of novel inhibitors of this pathway. In a cell-based high-throughput screening assay of 15,272 pure natural compounds, we identified pomiferin triacetate as a potent stabilizer of the tumor suppressor programmed cell death 4 (Pdcd4). Mechanistically, pomiferin triacetate appeared as a general inhibitor of the PI3K-Akt-mTOR-p70(S6K) cascade. Interference with this pathway occurred downstream of Akt but upstream of p70(S6K). Specifically, mTOR kinase emerged as the molecular target of pomiferin triacetate, with similar activities against mTOR complexes 1 and 2. In an in vitro mTOR kinase assay pomiferin triacetate dose-dependently inhibited mTOR with an IC50 of 6.2 μM. Molecular docking studies supported the interaction of the inhibitor with the catalytic site of mTOR. Importantly, pomiferin triacetate appeared to be highly selective for mTOR compared to a panel of 17 lipid and 50 protein kinases tested. As a consequence of the mTOR inhibition, pomiferin triacetate efficiently attenuated translation. In summary, pomiferin triacetate emerged as a novel and highly specific mTOR inhibitor with strong translation inhibitory effects. Thus, it might be an interesting lead structure for the development of mTOR- and translation-targeted anti-tumor therapies., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
34. PIF-pocket as a target for C. albicans Pkh selective inhibitors.
- Author
-
Pastor-Flores D, Schulze JO, Bahí A, Giacometti R, Ferrer-Dalmau J, Passeron S, Engel M, Süss E, Casamayor A, and Biondi RM
- Subjects
- Allosteric Regulation, Antifungal Agents chemistry, Antifungal Agents pharmacology, Binding Sites, Candida albicans enzymology, Chalcones chemistry, Enzyme Activation drug effects, Humans, Protein Kinase Inhibitors chemistry, Protein Kinases metabolism, Reactive Oxygen Species, Thioglycolates chemistry, Candida albicans drug effects, Chalcones pharmacology, Drug Delivery Systems, Models, Molecular, Protein Kinase Inhibitors pharmacology, Protein Kinases chemistry, Receptors, Neurotransmitter metabolism, Thioglycolates pharmacology
- Abstract
The phosphoinositide-dependent protein kinase 1, PDK1, is a master kinase that phosphorylates the activation loop of up to 23 AGC kinases. S. cerevisiae has three PDK1 orthologues, Pkh1-3, which also phosphorylate AGC kinases (e.g., Ypk, Tpk, Pkc1, and Sch9). Pkh1 and 2 are redundant proteins involved in multiple essential cellular functions, including endocytosis and cell wall integrity. Based on similarities with the budding yeast, the Pkh of fungal infectious species was postulated as a novel target for antifungals. Here, we found that depletion of Pkh eventually induces oxidative stress and DNA double-strand breaks, leading to programmed cell death. This finding supports Pkh as an antifungal target since pharmacological inhibition of Pkh would lead to the death of yeast cells, the ultimate goal of antifungals. It was therefore of interest to further investigate the possibility to develop Pkh inhibitors with selectivity for Candida Pkh that would not inhibit the human ortholog. Here, we describe C. albicans Pkh2 biochemically, structurally and by using chemical probes in comparison to human PDK1. We found that a regulatory site on the C. albicans Pkh2 catalytic domain, the PIF-pocket, diverges from human PDK1. Indeed, we identified and characterized PS77, a new small allosteric inhibitor directed to the PIF-pocket, which has increased selectivity for C. albicans Pkh2. Together, our results describe novel features of the biology of Pkh and chemical biology approaches that support the validation of Pkh as a drug target for selective antifungals.
- Published
- 2013
- Full Text
- View/download PDF
35. Differential stability of cell-free circulating microRNAs: implications for their utilization as biomarkers.
- Author
-
Köberle V, Pleli T, Schmithals C, Augusto Alonso E, Haupenthal J, Bönig H, Peveling-Oberhag J, Biondi RM, Zeuzem S, Kronenberger B, Waidmann O, and Piiper A
- Subjects
- Enzyme Inhibitors pharmacology, Healthy Volunteers, Humans, RNA Stability, Reverse Transcriptase Polymerase Chain Reaction, Ribonuclease, Pancreatic antagonists & inhibitors, Biomarkers, Tumor blood, Erythrocytes metabolism, MicroRNAs blood, MicroRNAs chemistry, Serum metabolism
- Abstract
Background: MicroRNAs circulating in the blood, stabilized by complexation with proteins and/or additionally by encapsulation in lipid vesicles, are currently being evaluated as biomarkers. The consequences of their differential association with lipids/vesicles for their stability and use as biomarkers are largely unexplored and are subject of the present study., Methods: The levels of a set of selected microRNAs were determined by quantitative reverse-transcription PCR after extraction from sera or vesicle- and non-vesicle fractions prepared from sera. The stability of these microRNAs after incubation with RNase A or RNase inhibitor, an inhibitor of RNase A family enzymes was studied., Results: The levels of microRNA-1 and microRNA-122, but not those of microRNA-16, microRNA-21 and microRNA-142-3p, declined significantly during a 5-h incubation of the sera. RNase inhibitor prevented the loss of microRNAs in serum as well as the degradation of microRNA-122, a microRNA not expressed in blood cells, in whole blood. Stabilization of microRNA-122 was also achieved by hemolysis. Prolonged incubation of the sera led to enrichment of vesicle-associated relative to non-vesicle-associated microRNAs. Vesicle-associated microRNAs were more resistant to RNase A treatment than the respective microRNAs not associated with vesicles., Conclusions: Serum microRNAs showed differential stability upon prolonged incubation. RNase inhibitor might be useful to robustly preserve the pattern of cell-free circulating microRNAs. In the case of microRNAs not expressed in blood cells this can also be achieved by hemolysis. Vesicle-associated microRNAs appeared to be more stable than those not associated with vesicles, which might be useful to disclose additional biomarker properties of miRNAs.
- Published
- 2013
- Full Text
- View/download PDF
36. AGC protein kinases: from structural mechanism of regulation to allosteric drug development for the treatment of human diseases.
- Author
-
Arencibia JM, Pastor-Flores D, Bauer AF, Schulze JO, and Biondi RM
- Subjects
- Allosteric Regulation, Candida albicans drug effects, Candida albicans enzymology, Candidiasis drug therapy, Candidiasis microbiology, Humans, Neoplasms drug therapy, Neoplasms enzymology, Phosphorylation drug effects, Protein Kinase Inhibitors pharmacology, Protein Kinases classification, Models, Molecular, Protein Kinases chemistry, Protein Kinases metabolism, Protein Structure, Tertiary
- Abstract
The group of AGC protein kinases includes more than 60 protein kinases in the human genome, classified into 14 families: PDK1, AKT/PKB, SGK, PKA, PKG, PKC, PKN/PRK, RSK, NDR, MAST, YANK, DMPK, GRK and SGK494. This group is also widely represented in other eukaryotes, including causative organisms of human infectious diseases. AGC kinases are involved in diverse cellular functions and are potential targets for the treatment of human diseases such as cancer, diabetes, obesity, neurological disorders, inflammation and viral infections. Small molecule inhibitors of AGC kinases may also have potential as novel therapeutic approaches against infectious organisms. Fundamental in the regulation of many AGC kinases is a regulatory site termed the "PIF-pocket" that serves as a docking site for substrates of PDK1. This site is also essential to the mechanism of activation of AGC kinases by phosphorylation and is involved in the allosteric regulation of N-terminal domains of several AGC kinases, such as PKN/PRKs and atypical PKCs. In addition, the C-terminal tail and its interaction with the PIF-pocket are involved in the dimerization of the DMPK family of kinases and may explain the molecular mechanism of allosteric activation of GRKs by GPCR substrates. In this review, we briefly introduce the AGC kinases and their known roles in physiology and disease and the discovery of the PIF-pocket as a regulatory site in AGC kinases. Finally, we summarize the current status and future therapeutic potential of small molecules directed to the PIF-pocket; these molecules can allosterically activate or inhibit the kinase as well as act as substrate-selective inhibitors. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases (2012)., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. InterAKTions with FKBPs--mutational and pharmacological exploration.
- Author
-
Fabian AK, März A, Neimanis S, Biondi RM, Kozany C, and Hausch F
- Subjects
- Allosteric Site, Phosphorylation, Protein Binding, Proto-Oncogene Proteins c-akt metabolism, Tacrolimus Binding Proteins genetics, Mutation, Tacrolimus Binding Proteins metabolism
- Abstract
The FK506-binding protein 51 (FKBP51) is an Hsp90-associated co-chaperone which regulates steroid receptors and kinases. In pancreatic cancer cell lines, FKBP51 was shown to recruit the phosphatase PHLPP to facilitate dephosphorylation of the kinase Akt, which was associated with reduced chemoresistance. Here we show that in addition to FKBP51 several other members of the FKBP family bind directly to Akt. FKBP51 can also form complexes with other AGC kinases and mapping studies revealed that FKBP51 interacts with Akt via multiple domains independent of their activation or phosphorylation status. The FKBP51-Akt1 interaction was not affected by FK506 analogs or Akt active site inhibitors, but was abolished by the allosteric Akt inhibitor VIII. None of the FKBP51 inhibitors affected AktS473 phosphorylation or downstream targets of Akt. In summary, we show that FKBP51 binds to Akt directly as well as via Hsp90. The FKBP51-Akt interaction is sensitive to the conformation of Akt1, but does not depend on the FK506-binding pocket of FKBP51. Therefore, FKBP inhibitors are unlikely to inhibit the Akt-FKBP-PHLPP network.
- Published
- 2013
- Full Text
- View/download PDF
38. 2-(3-Oxo-1,3-diphenylpropyl)malonic acids as potent allosteric ligands of the PIF pocket of phosphoinositide-dependent kinase-1: development and prodrug concept.
- Author
-
Wilhelm A, Lopez-Garcia LA, Busschots K, Fröhner W, Maurer F, Boettcher S, Zhang H, Schulze JO, Biondi RM, and Engel M
- Subjects
- Allosteric Site, Binding Sites, DNA Helicases metabolism, Enzyme Activation, Humans, Immunoblotting, Malonates chemical synthesis, Models, Molecular, Molecular Structure, Muscle Cells metabolism, Prodrugs chemical synthesis, Protein Binding, Protein Kinase Inhibitors chemical synthesis, Protein Serine-Threonine Kinases metabolism, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Structure-Activity Relationship, DNA Helicases antagonists & inhibitors, Drug Design, Malonates chemistry, Malonates pharmacology, Muscle Cells drug effects, Prodrugs pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
The protein kinase C-related kinase 2 (PRK2)-interacting fragment (PIF) pocket of phosphoinositide-dependent kinase-1 (PDK1) was proposed as a novel target site for allosteric modulators. In the present work, we describe the design, synthesis, and structure-activity relationship of a series of 2-(3-oxo-1,3-diphenylpropyl)malonic acids as potent allosteric activators binding to the PIF pocket. Some congeners displayed AC(50) values for PDK1 activation in the submicromolar range. The potency of the best compounds to stabilize PDK1 in a thermal stability shift assay was in the same order of magnitude as that of the PIF pocket binding peptide PIFtide, suggesting comparable binding affinities to the PIF pocket. The crystal structure of PDK1 in complex with compound 4h revealed that additional ionic interactions are mainly responsible for the increased potency compared to the monocarboxylate analogues. Notably, several compounds displayed high selectivity for PDK1. Employing a prodrug strategy, we were able to corroborate the novel mechanism of action in cells.
- Published
- 2012
- Full Text
- View/download PDF
39. Substrate-selective inhibition of protein kinase PDK1 by small compounds that bind to the PIF-pocket allosteric docking site.
- Author
-
Busschots K, Lopez-Garcia LA, Lammi C, Stroba A, Zeuzem S, Piiper A, Alzari PM, Neimanis S, Arencibia JM, Engel M, Schulze JO, and Biondi RM
- Subjects
- Animals, Cell Line, Chalcones chemistry, Dicarboxylic Acids chemistry, HEK293 Cells, Humans, Mice, Models, Biological, Models, Molecular, Molecular Structure, Molecular Weight, Prodrugs chemistry, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases metabolism, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Structure-Activity Relationship, Substrate Specificity, Allosteric Site drug effects, Chalcones pharmacology, Dicarboxylic Acids pharmacology, Prodrugs pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
The PIF-pocket of AGC protein kinases participates in the physiologic mechanism of regulation by acting as a docking site for substrates and as a switch for the transduction of the conformational changes needed for activation or inhibition. We describe the effects of compounds that bind to the PIF-pocket of PDK1. In vitro, PS210 is a potent activator of PDK1, and the crystal structure of the PDK1-ATP-PS210 complex shows that PS210 stimulates the closure of the kinase domain. However, in cells, the prodrug of PS210 (PS423) acts as a substrate-selective inhibitor of PDK1, inhibiting the phosphorylation and activation of S6K, which requires docking to the PIF-pocket, but not affecting PKB/Akt. This work describes a tool to study the dynamics of PDK1 activity and a potential approach for drug discovery., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
40. Regulation of protein kinase C-related protein kinase 2 (PRK2) by an intermolecular PRK2-PRK2 interaction mediated by Its N-terminal domain.
- Author
-
Bauer AF, Sonzogni S, Meyer L, Zeuzem S, Piiper A, Biondi RM, and Neimanis S
- Subjects
- Enzyme Activation physiology, HEK293 Cells, Humans, Phosphorylation physiology, Protein Kinase C genetics, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Protein Structure, Secondary, Protein Structure, Tertiary, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Protein Kinase C metabolism, Protein Multimerization physiology
- Abstract
Protein kinase C-related protein kinases (PRKs) are effectors of the Rho family of small GTPases and play a role in the development of diseases such as prostate cancer and hepatitis C. Here we examined the mechanism underlying the regulation of PRK2 by its N-terminal region. We show that the N-terminal region of PRK2 prevents the interaction with its upstream kinase, the 3-phosphoinositide-dependent kinase 1 (PDK1), which phosphorylates the activation loop of PRK2. We confirm that the N-terminal region directly inhibits the kinase activity of PRK2. However, in contrast to previous models, our data indicate that this inhibition is mediated in trans through an intermolecular PRK2-PRK2 interaction. Our results also suggest that amino acids 487-501, located in the linker region between the N-terminal domains and the catalytic domain, contribute to the PRK2-PRK2 dimer formation. This dimerization is further supported by other N-terminal domains. Additionally, we provide evidence that the region C-terminal to the catalytic domain intramolecularly activates PRK2. Finally, we discovered that the catalytic domain mediates a cross-talk between the inhibitory N-terminal region and the activating C-terminal region. The results presented here describe a novel mechanism of regulation among AGC kinases and offer new insights into potential approaches to pharmacologically regulate PRK2.
- Published
- 2012
- Full Text
- View/download PDF
41. Cross regulation between Candida albicans catalytic and regulatory subunits of protein kinase A.
- Author
-
Giacometti R, Kronberg F, Biondi RM, Hernández AI, and Passeron S
- Subjects
- Candida albicans genetics, Cyclic AMP-Dependent Protein Kinases genetics, Gene Deletion, Gene Expression Profiling, Mutant Proteins genetics, Mutant Proteins metabolism, Candida albicans enzymology, Candida albicans metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Gene Expression Regulation, Fungal
- Abstract
In the pathogen Candida albicans protein kinase A (PKA) catalytic subunit is encoded by two genes TPK1 and TPK2 and the regulatory subunit by one gene, BCY1. PKA mediates several cellular processes such as cell cycle regulation and the yeast to hyphae transition, a key factor for C. albicans virulence. The catalytic isoforms Tpk1p and Tpk2p share redundant functions in vegetative growth and hyphal development, though they differentially regulate glycogen metabolism, the stress response pathway and pseudohyphal formation. In Saccharomyces cerevisiae it was earlier reported that BCY1 overexpression not only increased the amount of TPK3 mRNA but also its catalytic activity. In C. albicans a significant decrease in Bcy1p expression levels was already observed in tpk2Δ null strains. In this work we showed that the upregulation in Bcy1p expression was observed in a set of strains having a TPK1 or TPK2 allele reintegrated in its own locus, as well as in strains expressing the TPKs under the control of the constitutive ACT1 promoter. To confirm the cross regulation event between Bcy1p and Tpkp expression we generated a mutant strain with the lowest PKA activity carrying one TPK1 and a unique BCY1 allele with the aim to obtain two derived strains in which BCY1 or TPK1 were placed under their own promoters inserted in the RPS10 neutral locus. We found that placing one copy of BCY1 upregulated the levels of Tpk1p and its catalytic activity; while TPK1 insertion led to an increase in BCY1 mRNA, Bcy1p and in a high cAMP binding activity. Our results suggest that C. albicans cells were able to compensate for the increased levels of either Tpk1p or Tpk2p subunits with a corresponding elevation of Bcy1 protein levels and vice versa, implying a tightly regulated mechanism to balance holoenzyme formation., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
42. Use of a fluorescent ATP analog to probe the allosteric conformational change in the active site of the protein kinase PDK1.
- Author
-
Hindie V, Lopez-Garcia LA, and Biondi RM
- Subjects
- 3-Phosphoinositide-Dependent Protein Kinases, Allosteric Site, Binding Sites, Catalytic Domain, Protein Conformation, Adenosine Triphosphate analogs & derivatives, Adenosine Triphosphate metabolism, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism
- Abstract
There is growing interest in exploring allosteric sites on proteins for drug discovery. At the center of the regulation of many protein kinases from the AGC family there is an allosteric site termed "PIF-pocket." The regulated binding of a C-terminal region of the kinase to the PIF-pocket, within the small lobe of the catalytic core, modulates the activity of AGC kinases. Small compounds that bind to the PIF-pocket can mimic its physiological mechanism of regulation and modulate the kinase activity in vitro, e.g., small compounds can activate the phosphoinositide-dependent protein kinase 1 (PDK1). Compounds binding to an allosteric site on a protein kinase may produce conformational changes at the ATP-binding site within the active site of the kinase domain. We here describe a fluorescent method using the ATP analog TNP-ATP that allows evaluating the allosteric conformational changes at the ATP-binding site of PDK1 triggered by small compounds binding to the PIF-pocket.
- Published
- 2012
- Full Text
- View/download PDF
43. Brain specific kinase-1 BRSK1/SAD-B associates with lipid rafts: modulation of kinase activity by lipid environment.
- Author
-
Rodríguez-Asiain A, Ruiz-Babot G, Romero W, Cubí R, Erazo T, Biondi RM, Bayascas JR, Aguilera J, Gómez N, Gil C, Claro E, and Lizcano JM
- Subjects
- AMP-Activated Protein Kinase Kinases, Animals, Baculoviridae, Escherichia coli, Fetus, HEK293 Cells, Humans, Lipoylation, Membranes, Artificial, Mice, Phosphorylation, Protein Structure, Secondary, Rats, Rats, Sprague-Dawley, Recombinant Proteins genetics, Threonine metabolism, Brain physiology, Intracellular Signaling Peptides and Proteins metabolism, Membrane Microdomains metabolism, Protein Serine-Threonine Kinases metabolism, Recombinant Proteins metabolism, Signal Transduction physiology, Synaptic Transmission physiology, Synaptosomes metabolism
- Abstract
Brain specific kinases 1 and 2 (BRSK1/2, also named SAD kinases) are serine-threonine kinases specifically expressed in the brain, and activated by LKB1-mediated phosphorylation of a threonine residue at their T-loop (Thr189/174 in human BRSK1/2). BRSKs are crucial for establishing neuronal polarity, and BRSK1 has also been shown to regulate neurotransmitter release presynaptically. How BRSK1 exerts this latter function is unknown, since its substrates at the synaptic terminal and the mechanisms modulating its activity remain to be described. Key regulators of neurotransmitter release, such as SNARE complex proteins, are located at membrane rafts. Therefore we initially undertook this work to check whether BRSK1 also locates at these membrane microdomains. Here we show that brain BRSK1, but not BRSK2, is palmitoylated, and provide biochemical and pharmacological evidences demonstrating that a pool of BRSK1, but not BRSK2 or LKB1, localizes at membrane lipid rafts. We also show that raft-associated BRSK1 has higher activity than BRSK1 from non-raft environment, based on a higher T-loop phosphorylation at Thr-189. Further, recombinant BRSK1 activity increased 3-fold when assayed with small multilamellar vesicles (SMV) generated with lipids extracted from synaptosomal raft fractions. A similar BRSK1-activating effect was obtained with synthetic SMV made with phosphatidylcholine, cholesterol and sphingomyelin, mixed in the same molar ratio at which these three major lipids are present in rafts. Importantly, SMV also enhanced the activity of a constitutively active BRSK1 (T189E), underpinning that interaction with lipid rafts represents a new mechanism of BRSK1 activity modulation, additional to T-loop phosphorylation., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
44. Allosteric regulation of protein kinase PKCζ by the N-terminal C1 domain and small compounds to the PIF-pocket.
- Author
-
Lopez-Garcia LA, Schulze JO, Fröhner W, Zhang H, Süss E, Weber N, Navratil J, Amon S, Hindie V, Zeuzem S, Jørgensen TJ, Alzari PM, Neimanis S, Engel M, and Biondi RM
- Subjects
- Allosteric Regulation drug effects, Binding Sites, Biocatalysis, Cell Line, Tumor, Humans, NF-kappa B metabolism, Protein Kinase C metabolism, Protein Structure, Tertiary, Signal Transduction, Small Molecule Libraries pharmacology, Protein Kinase C chemistry, Small Molecule Libraries chemistry
- Abstract
Protein kinases are key mediators of cellular signaling, and therefore, their activities are tightly controlled. AGC kinases are regulated by phosphorylation and by N- and C-terminal regions. Here, we studied the molecular mechanism of inhibition of atypical PKCζ and found that the inhibition by the N-terminal region cannot be explained by a simple pseudosubstrate inhibitory mechanism. Notably, we found that the C1 domain allosterically inhibits PKCζ activity and verified an allosteric communication between the PIF-pocket of atypical PKCs and the binding site of the C1 domain. Finally, we developed low-molecular-weight compounds that bind to the PIF-pocket and allosterically inhibit PKCζ activity. This work establishes a central role for the PIF-pocket on the regulation of PKCζ and allows us to envisage development of drugs targeting the PIF-pocket that can either activate or inhibit AGC kinases., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
45. 4-benzimidazolyl-3-phenylbutanoic acids as novel PIF-pocket-targeting allosteric inhibitors of protein kinase PKCζ.
- Author
-
Fröhner W, Lopez-Garcia LA, Neimanis S, Weber N, Navratil J, Maurer F, Stroba A, Zhang H, Biondi RM, and Engel M
- Subjects
- 3-Phosphoinositide-Dependent Protein Kinases, Allosteric Regulation, Binding Sites, Butyrates chemistry, Butyrates pharmacology, Cell-Free System, Humans, NF-kappa B metabolism, Protein Binding, Protein Serine-Threonine Kinases metabolism, Recombinant Proteins antagonists & inhibitors, Signal Transduction drug effects, Stereoisomerism, Structure-Activity Relationship, U937 Cells, Butyrates chemical synthesis, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism
- Abstract
Protein kinase inhibitors with an allosteric mode of action are expected to reach, in many cases, higher selectivity for the target enzyme than ATP-competitive compounds. Therefore, basic research is aiming at identifying and establishing novel sites on the catalytic domain of protein kinases which might be targeted by allosteric inhibitors. We previously published the first structure-activity relationships (SARs) for allosteric activators of protein kinase PDK1. Here, we present the design, synthesis, and SAR data on a series of novel compounds, 4-benzimidazolyl-3-phenylbutanoic acids, that inhibit the atypical protein kinace C (PKC) ζ via binding to the PIF-pocket. Key positions were identified in the compounds that can be modified to increase potency and selectivity. Some congeners showed a high selectivity toward PKCζ, lacking inhibition of the most closely related isoform, PKCι, and of further AGC kinases. Furthermore, evidence is provided that these compounds are also active toward cellular PKCζ without loss of potency compared to the cell-free assay.
- Published
- 2011
- Full Text
- View/download PDF
46. Candida albicans Tpk1p and Tpk2p isoforms differentially regulate pseudohyphal development, biofilm structure, cell aggregation and adhesins expression.
- Author
-
Giacometti R, Kronberg F, Biondi RM, and Passeron S
- Subjects
- Candida albicans enzymology, Candida albicans genetics, Candida albicans physiology, Cell Adhesion, Cyclic AMP-Dependent Protein Kinases chemistry, Cyclic AMP-Dependent Protein Kinases genetics, Fungal Proteins chemistry, Fungal Proteins genetics, Gene Expression Regulation, Fungal, Hyphae chemistry, Hyphae enzymology, Hyphae genetics, Hyphae growth & development, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Biofilms, Candida albicans growth & development, Cyclic AMP-Dependent Protein Kinases metabolism, Fungal Proteins metabolism, Gene Expression Regulation, Developmental
- Abstract
Candida albicans undergoes a reversible morphological transition from single yeast cells to pseudohyphal and hyphal filaments. In this organism, cAMP-dependent protein kinase (PKA), coded by two catalytic subunits (TPK1 and TPK2) and one regulatory subunit (BCY1), mediates basic cellular processes, such as the yeast-to-hypha transition and cell cycle regulation. It is known that both Tpk isoforms play positive roles in vegetative growth and filamentation, although distinct roles have been found in virulence, stress response and glycogen storage. However, little is known regarding the participation of Tpk1p and/or Tpk2p in pseudohyphal development. This point was addressed using several C. albicans PKA mutants having heterozygous or homozygous deletions of TPK1 and/or TPK2 in different BCY1 genetic backgrounds. We observed that under hypha-only inducing conditions, all BCY1 heterozygous strains shifted growth toward pseudohyphal morphology; however, the pseudohypha:hypha ratio was higher in strains devoid of TPK2. Under pseudohypha-only inducing conditions, strains lacking TPK2 were prone to develop short and branched pseudohyphae. In tpk2 Δ/tpk2 Δ strains, biofilm architecture was markedly less dense, composed of short pseudohyphae and blastospores with reduced adhesion ability to abiotic material, suggesting a significant defect in cell adherence. Immunolabelling assays showed a decreased expression of adhesins Als1p and Als3p only in the tpk2 Δ/tpk2 Δ strain. Complementation of this mutant with a wild-type copy of TPK2 restored all the altered functions: pseudohyphae elongation, biofilm composition, cell aggregation and adhesins expression. Our study suggests that the Tpk2p isoform may be part of a mechanism underlying not only polarized pseudohyphal morphogenesis but also cell adherence., (Copyright © 2011 John Wiley & Sons, Ltd.)
- Published
- 2011
- Full Text
- View/download PDF
47. Inhibition of the equilibrative nucleoside transporter 1 and activation of A2A adenosine receptors by 8-(4-chlorophenylthio)-modified cAMP analogs and their hydrolytic products.
- Author
-
Waidmann O, Pleli T, Dvorak K, Baehr C, Mondorf U, Plotz G, Biondi RM, Zeuzem S, and Piiper A
- Subjects
- Amino Acid Motifs, Animals, Apoptosis, Cell Line, Coloring Agents pharmacology, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases chemistry, Humans, Hydrolysis, PC12 Cells, Phosphorylation, Rats, Tetrazolium Salts pharmacology, Thiazoles pharmacology, Cyclic AMP chemistry, Equilibrative Nucleoside Transporter 1 metabolism, Receptor, Adenosine A2A chemistry
- Abstract
Cyclic AMP analogs containing hydrophobic modification of C(8) at the adenine ring such as 8-(4-chlorophenylthio)-cAMP (8-pCPT-cAMP) and 8-(4-chlorophenylthio)-2'-O-methyl-cAMP (8-pCPT-2'-O-methyl-cAMP) can penetrate membranes due to their high lipophilicity and directly activate intracellular cAMP effectors. Therefore, these cAMP analogs have been used in numerous studies, assuming that their effects reflect the consequences of direct activation of cAMP effectors. The present study provides evidence that 8-pCPT-modified cAMP analogs and their corresponding putative hydrolysis products (8-(4-chlorophenylthio)-adenosine (8-pCPT-ado) and 8-(4-chlorophenylthio)-2'-O-methyl-adenosine (8-pCPT-2'-O-methyl-ado)) inhibit the equilibrative nucleoside transporter 1 (ENT1). In PC12 cells, in which nucleoside transport strongly depended on ENT1, 8-pCPT-ado, 8-pCPT-2'-O-methyl-ado, and, to a smaller extent, 8-pCPT-2'-O-methyl-cAMP caused an increase of protein kinase A substrate motif phosphorylation and anti-apoptotic effect by an A(2A) adenosine receptor (A(2A)R)-dependent mechanism. In contrast, the effects of 8-pCPT-cAMP were mainly A(2A)R-independent. In HEK 293 showing little endogenous ENT1-dependent nucleoside transport, transfection of ENT1 conferred A(2A)R-dependent increase in protein kinase A substrate motif phosphorylation. Together, the data of the present study indicate that inhibition of ENT1 and activation of adenosine receptors have to be considered when interpreting the effects of 8-pCPT-substituted cAMP/adenosine analogs.
- Published
- 2009
- Full Text
- View/download PDF
48. Regulation of the interaction between protein kinase C-related protein kinase 2 (PRK2) and its upstream kinase, 3-phosphoinositide-dependent protein kinase 1 (PDK1).
- Author
-
Dettori R, Sonzogni S, Meyer L, Lopez-Garcia LA, Morrice NA, Zeuzem S, Engel M, Piiper A, Neimanis S, Frödin M, and Biondi RM
- Subjects
- 3-Phosphoinositide-Dependent Protein Kinases, Binding Sites, Cell Line, Humans, Models, Molecular, Phosphorylation, Protein Binding, Protein Kinase C metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
The members of the AGC kinase family frequently exhibit three conserved phosphorylation sites: the activation loop, the hydrophobic motif (HM), and the zipper (Z)/turn-motif (TM) phosphorylation site. 3-Phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylates the activation loop of numerous AGC kinases, including the protein kinase C-related protein kinases (PRKs). Here we studied the docking interaction between PDK1 and PRK2 and analyzed the mechanisms that regulate this interaction. In vivo labeling of recombinant PRK2 by (32)P(i) revealed phosphorylation at two sites, the activation loop and the Z/TM in the C-terminal extension. We provide evidence that phosphorylation of the Z/TM site of PRK2 inhibits its interaction with PDK1. Our studies further provide a mechanistic model to explain different steps in the docking interaction and regulation. Interestingly, we found that the mechanism that negatively regulates the docking interaction of PRK2 to the upstream kinase PDK1 is directly linked to the activation mechanism of PRK2 itself. Finally, our results indicate that the mechanisms underlying the regulation of the interaction between PRK2 and PDK1 are specific for PRK2 and do not apply for other AGC kinases.
- Published
- 2009
- Full Text
- View/download PDF
49. Structure and allosteric effects of low-molecular-weight activators on the protein kinase PDK1.
- Author
-
Hindie V, Stroba A, Zhang H, Lopez-Garcia LA, Idrissova L, Zeuzem S, Hirschberg D, Schaeffer F, Jørgensen TJ, Engel M, Alzari PM, and Biondi RM
- Subjects
- 3-Phosphoinositide-Dependent Protein Kinases, Allosteric Regulation, Allosteric Site, Cell Line, Crystallography, X-Ray, Enzyme Activation, Humans, Models, Molecular, Molecular Weight, Phosphorylation, Protein Conformation, Protein Serine-Threonine Kinases genetics, Substrate Specificity, Phosphopeptides pharmacology, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism
- Abstract
Protein phosphorylation transduces a large set of intracellular signals. One mechanism by which phosphorylation mediates signal transduction is by prompting conformational changes in the target protein or interacting proteins. Previous work described an allosteric site mediating phosphorylation-dependent activation of AGC kinases. The AGC kinase PDK1 is activated by the docking of a phosphorylated motif from substrates. Here we present the crystallography of PDK1 bound to a rationally developed low-molecular-weight activator and describe the conformational changes induced by small compounds in the crystal and in solution using a fluorescence-based assay and deuterium exchange experiments. Our results indicate that the binding of the compound produces local changes at the target site, the PIF binding pocket, and also allosteric changes at the ATP binding site and the activation loop. Altogether, we present molecular details of the allosteric changes induced by small compounds that trigger the activation of PDK1 through mimicry of phosphorylation-dependent conformational changes.
- Published
- 2009
- Full Text
- View/download PDF
50. 3,5-Diphenylpent-2-enoic acids as allosteric activators of the protein kinase PDK1: structure-activity relationships and thermodynamic characterization of binding as paradigms for PIF-binding pocket-targeting compounds.
- Author
-
Stroba A, Schaeffer F, Hindie V, Lopez-Garcia L, Adrian I, Fröhner W, Hartmann RW, Biondi RM, and Engel M
- Subjects
- Allosteric Site, Calorimetry, Entropy, Fatty Acids, Monounsaturated chemistry, Fatty Acids, Monounsaturated metabolism, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Structure-Activity Relationship, Fatty Acids, Monounsaturated chemical synthesis, Protein Serine-Threonine Kinases drug effects, Thermodynamics
- Abstract
The modulation of protein kinase activities by low molecular weight compounds is a major goal of current pharmaceutical developments. In this line, important efforts are directed to the development of drugs targeting the conserved ATP binding site. However, there is very little experience on targeting allosteric, regulatory sites, different from the ATP binding site, in protein kinases. Here we describe the synthesis, cell-free activation potency, and calorimetric binding analysis of 3,5-diphenylpent-2-enoic acids and derivatives as allosteric modulators of the phosphoinositide-dependent kinase-1 (PDK1) catalytic activity. Our SAR results combined with thermodynamic binding analyses revealed both favorable binding enthalpy and entropy and confirmed the PIF-binding pocket of PDK1 as a druggable site. In conclusion, we defined the minimal structural requirements for compounds to bind to the PIF-binding pocket and to act as allosteric modulators and identified two new lead structures (12Z and 13Z) with predominating binding enthalpy.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.