Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Front Pharmacol ; 11: 615211, 2020.
Article in English | MEDLINE | ID: mdl-33381049

ABSTRACT

In-crystal fragment screening is a powerful tool to chemically probe the surfaces used by proteins to interact, and identify the chemical space worth exploring to design protein-protein inhibitors. A crucial prerequisite is the identification of a crystal form where the target area is exposed and accessible to be probed by fragments. Here we report a crystal form of the SARS-CoV-2 Receptor Binding Domain in complex with the CR3022 antibody where the ACE2 binding site on the Receptor Binding Domain is exposed and accessible. This crystal form of the complex is a valuable tool to develop antiviral molecules that could act by blocking the virus entry in cells.

2.
J Med Chem ; 63(14): 7559-7568, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32543856

ABSTRACT

Nowadays, it is possible to combine X-ray crystallography and fragment screening in a medium throughput fashion to chemically probe the surfaces used by proteins to interact and use the outcome of the screens to systematically design protein-protein inhibitors. To prove it, we first performed a bioinformatics analysis of the Protein Data Bank protein complexes, which revealed over 400 cases where the crystal lattice of the target in the free form is such that large portions of the interacting surfaces are free from lattice contacts and therefore accessible to fragments during soaks. Among the tractable complexes identified, we then performed single fragment crystal screens on two particular interesting cases: the Il1ß-ILR and p38α-TAB1 complexes. The result of the screens showed that fragments tend to bind in clusters, highlighting the small-molecule hotspots on the surface of the target protein. In most of the cases, the hotspots overlapped with the binding sites of the interacting proteins.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Interleukin-1beta/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Protein Multimerization/drug effects , Receptors, Interleukin-1/metabolism , Adamantane/analogs & derivatives , Adamantane/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Databases, Protein , Humans , Interleukin-1beta/chemistry , Mitogen-Activated Protein Kinase 14/chemistry , Protein Binding/drug effects , Receptors, Interleukin-1/chemistry , Sulfonamides/chemistry , Sulfonamides/metabolism , Yeasts/chemistry
3.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135318

ABSTRACT

Inhibiting MAPK14 (p38α) diminishes cardiac damage in myocardial ischemia. During myocardial ischemia, p38α interacts with TAB1, a scaffold protein, which promotes p38α autoactivation; active p38α (pp38α) then transphosphorylates TAB1. Previously, we solved the X-ray structure of the p38α-TAB1 (residues 384-412) complex. Here, we further characterize the interaction by solving the structure of the pp38α-TAB1 (residues 1-438) complex in the active state. Based on this information, we created a global knock-in (KI) mouse with substitution of 4 residues on TAB1 that we show are required for docking onto p38α. Whereas ablating p38α or TAB1 resulted in early embryonal lethality, the TAB1-KI mice were viable and had no appreciable alteration in their lymphocyte repertoire or myocardial transcriptional profile; nonetheless, following in vivo regional myocardial ischemia, infarction volume was significantly reduced and the transphosphorylation of TAB1 was disabled. Unexpectedly, the activation of myocardial p38α during ischemia was only mildly attenuated in TAB1-KI hearts. We also identified a group of fragments able to disrupt the interaction between p38α and TAB1. We conclude that the interaction between the 2 proteins can be targeted with small molecules. The data reveal that it is possible to selectively inhibit signaling downstream of p38α to attenuate ischemic injury.


Subject(s)
Adamantane/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Adamantane/analogs & derivatives , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Crystallography, X-Ray , Disease Models, Animal , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/genetics , Mutation , Myocardial Infarction/drug therapy , Myocardial Infarction/etiology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Reperfusion Injury/complications
4.
Mol Cell Biol ; 38(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29229647

ABSTRACT

p38α mitogen-activated protein kinase is essential to cellular homeostasis. Two principal mechanisms to activate p38α exist. The first relies on dedicated dual-specificity kinases such as mitogen-activated protein kinase kinase (MAP2K) 3 (MKK3) or 6 (MKK6), which activate p38α by phosphorylating Thr180 and Tyr182 within the activation segment. The second is by autophosphorylation of Thr180 and Tyr182 in cis, mediated by p38α binding the scaffold protein TAB1. The second mechanism occurs during myocardial ischemia, where it aggravates myocardial infarction. Based on the crystal structure of the p38α-TAB1 complex we replaced threonine 185 of p38α with glycine (T185G) to prevent an intramolecular hydrogen bond with Asp150 from being formed. This mutation did not interfere with TAB1 binding to p38α. However, it disrupted the consequent long-range effect of this binding event on the distal activation segment, releasing the constraint on Thr180 that oriented its hydroxyl for phosphotransfer. Based on assays performed in vitro and in vivo, the autoactivation of p38α(T185G) was disabled, while its ability to be activated by upstream MAP2Ks and to phosphorylate downstream substrates remained intact. Furthermore, myocardial cells expressing p38α(T185G) were resistant to injury. These findings reveal a mechanism to selectively disable p38α autoactivation and its consequences, which may ultimately circumvent the toxicity associated with strategies that inhibit p38α kinase activity under all circumstances, such as with ATP-competitive inhibitors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Amino Acid Sequence , Binding Sites , Enzyme Activation , HEK293 Cells , Humans , MAP Kinase Kinase 3/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Threonine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL