Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Chem Biol ; 16(7): 1191-1200, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34161732

ABSTRACT

Intrinsically disordered regions in proteins often function as binding motifs in protein-protein interactions. The mechanistic aspects and molecular details of such coupled binding and folding reactions, which involve formation of multiple noncovalent bonds, have been broadly studied theoretically, but experimental data are scarce. Here, using a combination of protein semisynthesis to incorporate phosphorylated amino acids, backbone amide-to-ester modifications, side chain substitutions, and binding kinetics, we examined the interaction between the intrinsically disordered motif of amyloid precursor protein (APP) and the phosphotyrosine binding (PTB) domain of Mint2. We show that the interaction is regulated by a self-inhibitory segment of the PTB domain previously termed ARM. The helical ARM linker decreases the association rate constant 30-fold through a fast pre-equilibrium between an open and a closed state. Extensive side chain substitutions combined with kinetic experiments demonstrate that the rate-limiting transition state for the binding reaction is governed by native and non-native hydrophobic interactions and hydrogen bonds. Hydrophobic interactions were found to be particularly important during crossing of the transition state barrier. Furthermore, linear free energy relationships show that the overall coupled binding and folding reaction involves cooperative formation of interactions with roughly 30% native contacts formed at the transition state. Our data support an emerging picture of coupled binding and folding reactions following overall chemical principles similar to those of folding of globular protein domains but with greater malleability of ground and transition states.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cadherins/metabolism , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Amyloid beta-Protein Precursor/chemical synthesis , Amyloid beta-Protein Precursor/genetics , Animals , Cadherins/chemical synthesis , Cadherins/genetics , Carrier Proteins/chemical synthesis , Carrier Proteins/genetics , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Intrinsically Disordered Proteins/chemical synthesis , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Kinetics , Mutation , Nerve Tissue Proteins/chemical synthesis , Nerve Tissue Proteins/genetics , Protein Binding , Protein Domains/genetics , Protein Engineering , Protein Folding , Rats , Thermodynamics
2.
J Am Chem Soc ; 143(2): 891-901, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33398998

ABSTRACT

There is an urgent need for novel therapeutic approaches to treat Alzheimer's disease (AD) with the ability to both alleviate the clinical symptoms and halt the progression of the disease. AD is characterized by the accumulation of amyloid-ß (Aß) peptides which are generated through the sequential proteolytic cleavage of the amyloid precursor protein (APP). Previous studies reported that Mint2, a neuronal adaptor protein binding both APP and the γ-secretase complex, affects APP processing and formation of pathogenic Aß. However, there have been contradicting results concerning whether Mint2 has a facilitative or suppressive effect on Aß generation. Herein, we deciphered the APP-Mint2 protein-protein interaction (PPI) via extensive probing of both backbone H-bond and side-chain interactions. We also developed a proteolytically stable, high-affinity peptide targeting the APP-Mint2 interaction. We found that both an APP binding-deficient Mint2 variant and a cell-permeable PPI inhibitor significantly reduced Aß42 levels in a neuronal in vitro model of AD. Together, these findings demonstrate a facilitative role of Mint2 in Aß formation, and the combination of genetic and pharmacological approaches suggests that targeting Mint2 is a promising therapeutic strategy to reduce pathogenic Aß levels.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Cadherins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Peptides/pharmacology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cadherins/metabolism , Humans , Nerve Tissue Proteins/metabolism , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding/drug effects
3.
J Med Chem ; 64(3): 1423-1434, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33502198

ABSTRACT

Despite the recent advances in cancer therapeutics, highly aggressive cancer forms, such as glioblastoma (GBM), still have very low survival rates. The intracellular scaffold protein syntenin, comprising two postsynaptic density protein-95/discs-large/zona occludens-1 (PDZ) domains, has emerged as a novel therapeutic target in highly malignant phenotypes including GBM. Here, we report the development of a novel, highly potent, and metabolically stable peptide inhibitor of syntenin, KSL-128114, which binds the PDZ1 domain of syntenin with nanomolar affinity. KSL-128114 is resistant toward degradation in human plasma and mouse hepatic microsomes and displays a global PDZ domain selectivity for syntenin. An X-ray crystal structure reveals that KSL-128114 interacts with syntenin PDZ1 in an extended noncanonical binding mode. Treatment with KSL-128114 shows an inhibitory effect on primary GBM cell viability and significantly extends survival time in a patient-derived xenograft mouse model. Thus, KSL-128114 is a novel promising candidate with therapeutic potential for highly aggressive tumors, such as GBM.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Peptides/chemistry , Peptides/pharmacology , Syntenins/drug effects , Animals , Cell Line, Tumor , Drug Delivery Systems , High-Throughput Screening Assays , Humans , Ligands , Mice , Microsomes/metabolism , Models, Molecular , Mutation , Protein Binding , X-Ray Diffraction , Xenograft Model Antitumor Assays
4.
Chembiochem ; 19(20): 2136-2145, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30073762

ABSTRACT

All proteins contain characteristic backbones formed of consecutive amide bonds, which can engage in hydrogen bonds. However, the importance of these is not easily addressed by conventional technologies that only allow for side-chain substitutions. By contrast, technologies such as nonsense suppression mutagenesis and protein ligation allow for manipulation of the protein backbone. In particular, replacing the backbone amide groups with ester groups, that is, amide-to-ester mutations, is a powerful tool to examine backbone-mediated hydrogen bonds. In this minireview, we showcase examples of how amide-to-ester mutations can be used to uncover pivotal roles of backbone-mediated hydrogen bonds in protein recognition, folding, function, and structure.


Subject(s)
Amides/chemistry , Codon, Nonsense , Esters/chemistry , Proteins/chemistry , Proteins/genetics , Hydrogen Bonding , Mutagenesis , Protein Conformation , Protein Folding
5.
Chembiochem ; 2018 Mar 26.
Article in English | MEDLINE | ID: mdl-29578633

ABSTRACT

The intracellular adaptor protein Mint2 binds amyloid precursor protein (APP) and presenilin-1, which are both central constituents of the amyloidogenic pathway associated with Alzheimer's disease (AD). Additional interaction partners have also been suggested for Mint2; several of them are also pertinent to AD pathogenesis. However, no comparative mapping of the Mint2 protein-protein interaction network is available. Here we provide a systematic characterization of seven interaction partners and address their specificities towards the different binding domains of Mint2, which reveal domain-specific and -nonspecific interaction partners. Moreover, we show that the last two C-terminal amino acids of Mint2 are both important for the intramolecular interaction with the PDZ1 domain and for the stability of Mint2.

SELECTION OF CITATIONS
SEARCH DETAIL