Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Publication year range
1.
Nature ; 629(8014): 1133-1141, 2024 May.
Article in English | MEDLINE | ID: mdl-38750368

ABSTRACT

The N-methyl-D-aspartate (NMDA) receptor is a glutamate-activated cation channel that is critical to many processes in the brain. Genome-wide association studies suggest that glutamatergic neurotransmission and NMDA receptor-mediated synaptic plasticity are important for body weight homeostasis1. Here we report the engineering and preclinical development of a bimodal molecule that integrates NMDA receptor antagonism with glucagon-like peptide-1 (GLP-1) receptor agonism to effectively reverse obesity, hyperglycaemia and dyslipidaemia in rodent models of metabolic disease. GLP-1-directed delivery of the NMDA receptor antagonist MK-801 affects neuroplasticity in the hypothalamus and brainstem. Importantly, targeting of MK-801 to GLP-1 receptor-expressing brain regions circumvents adverse physiological and behavioural effects associated with MK-801 monotherapy. In summary, our approach demonstrates the feasibility of using peptide-mediated targeting to achieve cell-specific ionotropic receptor modulation and highlights the therapeutic potential of unimolecular mixed GLP-1 receptor agonism and NMDA receptor antagonism for safe and effective obesity treatment.


Subject(s)
Dizocilpine Maleate , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Obesity , Receptors, N-Methyl-D-Aspartate , Animals , Humans , Male , Mice , Rats , Brain Stem/metabolism , Brain Stem/drug effects , Disease Models, Animal , Dizocilpine Maleate/adverse effects , Dizocilpine Maleate/pharmacology , Dizocilpine Maleate/therapeutic use , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Obesity/drug therapy , Obesity/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
2.
EMBO Rep ; 25(7): 3040-3063, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849673

ABSTRACT

Polarized vesicular trafficking directs specific receptors and ion channels to cilia, but the underlying mechanisms are poorly understood. Here we describe a role for DLG1, a core component of the Scribble polarity complex, in regulating ciliary protein trafficking in kidney epithelial cells. Conditional knockout of Dlg1 in mouse kidney causes ciliary elongation and cystogenesis, and cell-based proximity labeling proteomics and fluorescence microscopy show alterations in the ciliary proteome upon loss of DLG1. Specifically, the retromer-associated protein SDCCAG3, IFT20, and polycystin-2 (PC2) are reduced in the cilia of DLG1-deficient cells compared to control cells. This phenotype is recapitulated in vivo and rescuable by re-expression of wild-type DLG1, but not a Congenital Anomalies of the Kidney and Urinary Tract (CAKUT)-associated DLG1 variant, p.T489R. Finally, biochemical approaches and Alpha Fold modelling suggest that SDCCAG3 and IFT20 form a complex that associates, at least indirectly, with DLG1. Our work identifies a key role for DLG1 in regulating ciliary protein composition and suggests that ciliary dysfunction of the p.T489R DLG1 variant may contribute to CAKUT.


Subject(s)
Carrier Proteins , Cilia , Discs Large Homolog 1 Protein , TRPP Cation Channels , Animals , Cilia/metabolism , TRPP Cation Channels/metabolism , TRPP Cation Channels/genetics , Mice , Discs Large Homolog 1 Protein/metabolism , Carrier Proteins/metabolism , Carrier Proteins/genetics , Humans , Protein Transport , Mice, Knockout , Kidney/metabolism , Epithelial Cells/metabolism , Protein Binding , Vesico-Ureteral Reflux/metabolism , Vesico-Ureteral Reflux/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Urogenital Abnormalities
3.
Angew Chem Int Ed Engl ; 63(26): e202400350, 2024 06 21.
Article in English | MEDLINE | ID: mdl-38602024

ABSTRACT

Macrocycles offer an attractive format for drug development due to their good binding properties and potential to cross cell membranes. To efficiently identify macrocyclic ligands for new targets, methods for the synthesis and screening of large combinatorial libraries of small cyclic peptides were developed, many of them using thiol groups for efficient peptide macrocyclization. However, a weakness of these libraries is that invariant thiol-containing building blocks such as cysteine are used, resulting in a region that does not contribute to library diversity but increases molecule size. Herein, we synthesized a series of structurally diverse thiol-containing elements and used them for the combinatorial synthesis of a 2,688-member library of small, structurally diverse peptidic macrocycles with unprecedented skeletal complexity. We then used this library to discover potent thrombin and plasma kallikrein inhibitors, some also demonstrating favorable membrane permeability. X-ray structure analysis of macrocycle-target complexes showed that the size and shape of the newly developed thiol elements are key for binding. The strategy and library format presented in this work significantly enhance structural diversity by allowing combinatorial modifications to a previously invariant region of peptide macrocycles, which may be broadly applied in the development of membrane permeable therapeutics.


Subject(s)
Macrocyclic Compounds , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/chemical synthesis , Humans , Cell Membrane Permeability , Peptides, Cyclic/chemistry , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Molecular Structure , Small Molecule Libraries/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Small Molecule Libraries/metabolism , Thrombin/metabolism , Thrombin/antagonists & inhibitors , Thrombin/chemistry , Crystallography, X-Ray , Sulfhydryl Compounds/chemistry , Models, Molecular
4.
J Biol Chem ; 298(12): 102688, 2022 12.
Article in English | MEDLINE | ID: mdl-36370848

ABSTRACT

Parkinson's disease is a neurodegenerative movement disorder associated with the intracellular aggregation of α-synuclein (α-syn). Cytotoxicity is mainly associated with the oligomeric species (αSOs) formed at early stages in α-syn aggregation. Consequently, there is an intense focus on the discovery of novel inhibitors such as peptides to inhibit oligomer formation and toxicity. Here, using peptide arrays, we identified nine peptides with high specificity and affinity for αSOs. Of these, peptides p194, p235, and p249 diverted α-syn aggregation from fibrils to amorphous aggregates with reduced ß-structures and increased random coil content. However, they did not reduce αSO cytotoxicity and permeabilization of large anionic unilamellar vesicles. In parallel, we identified a non-self-aggregating peptide (p216), derived from the cell-penetrating peptide penetratin, which showed 12-fold higher binding affinity to αSOs than to α-syn monomers (Kdapp 2.7 and 31.2 µM, respectively). p216 reduced αSOs-induced large anionic unilamellar vesicle membrane permeability at 10-1 to 10-3 mg/ml by almost 100%, was not toxic to SH-SY5Y cells, and reduced αSOs cytotoxicity by about 20%. We conclude that p216 is a promising starting point from which to develop peptides targeting toxic αSOs in Parkinson's disease.


Subject(s)
Cell-Penetrating Peptides , Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , Cell-Penetrating Peptides/isolation & purification , Cell-Penetrating Peptides/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Cell Line, Tumor
5.
Proc Natl Acad Sci U S A ; 117(13): 7447-7454, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32165542

ABSTRACT

Acid-sensing ion channels (ASICs) are proton-gated cation channels that contribute to neurotransmission, as well as initiation of pain and neuronal death following ischemic stroke. As such, there is a great interest in understanding the in vivo regulation of ASICs, especially by endogenous neuropeptides that potently modulate ASICs. The most potent endogenous ASIC modulator known to date is the opioid neuropeptide big dynorphin (BigDyn). BigDyn is up-regulated in chronic pain and increases ASIC-mediated neuronal death during acidosis. Understanding the mechanism and site of action of BigDyn on ASICs could thus enable the rational design of compounds potentially useful in the treatment of pain and ischemic stroke. To this end, we employ a combination of electrophysiology, voltage-clamp fluorometry, synthetic BigDyn analogs, and noncanonical amino acid-mediated photocrosslinking. We demonstrate that BigDyn binding results in an ASIC1a closed resting conformation that is distinct from open and desensitized states induced by protons. Using alanine-substituted BigDyn analogs, we find that the BigDyn modulation of ASIC1a is primarily mediated through electrostatic interactions of basic amino acids in the BigDyn N terminus. Furthermore, neutralizing acidic amino acids in the ASIC1a extracellular domain reduces BigDyn effects, suggesting a binding site at the acidic pocket. This is confirmed by photocrosslinking using the noncanonical amino acid azidophenylalanine. Overall, our data define the mechanism of how BigDyn modulates ASIC1a, identify the acidic pocket as the binding site for BigDyn, and thus highlight this cavity as an important site for the development of ASIC-targeting therapeutics.


Subject(s)
Acid Sensing Ion Channels/metabolism , Dynorphins/metabolism , Acid Sensing Ion Channels/genetics , Animals , Animals, Genetically Modified , Binding Sites , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Neurons/metabolism , Neuropeptides/metabolism , Neuropeptides/physiology , Oocytes/metabolism , Protons , Xenopus laevis
6.
J Biol Chem ; 297(2): 100953, 2021 08.
Article in English | MEDLINE | ID: mdl-34270957

ABSTRACT

Phenol-soluble modulins (PSMs), such as α-PSMs, ß-PSMs, and δ-toxin, are virulence peptides secreted by different Staphylococcus aureus strains. PSMs are able to form amyloid fibrils, which may strengthen the biofilm matrix that promotes bacterial colonization of and extended growth on surfaces (e.g., cell tissue) and increases antibiotic resistance. Many components contribute to biofilm formation, including the human-produced highly sulfated glycosaminoglycan heparin. Although heparin promotes S. aureus infection, the molecular basis for this is unclear. Given that heparin is known to induce fibrillation of a wide range of proteins, we hypothesized that heparin aids bacterial colonization by promoting PSM fibrillation. Here, we address this hypothesis using a combination of thioflavin T-fluorescence kinetic studies, CD, FTIR, electron microscopy, and peptide microarrays to investigate the mechanism of aggregation, the structure of the fibrils, and identify possible binding regions. We found that heparin accelerates fibrillation of all α-PSMs (except PSMα2) and δ-toxin but inhibits ß-PSM fibrillation by blocking nucleation or reducing fibrillation levels. Given that S. aureus secretes higher levels of α-PSM than ß-PSM peptides, heparin is therefore likely to promote fibrillation overall. Heparin binding is driven by multiple positively charged lysine residues in α-PSMs and δ-toxins, the removal of which strongly reduced binding affinity. Binding of heparin did not affect the structure of the resulting fibrils, that is, the outcome of the aggregation process. Rather, heparin provided a scaffold to catalyze or inhibit fibrillation. Based on our findings, we speculate that heparin may strengthen the bacterial biofilm and therefore enhance colonization via increased PSM fibrillation.


Subject(s)
Peptides , Staphylococcus aureus , Bacterial Toxins , Biofilms/growth & development , Kinetics , Peptides/metabolism , Virulence
7.
J Biol Chem ; 296: 100473, 2021.
Article in English | MEDLINE | ID: mdl-33639163

ABSTRACT

The type II sodium-dependent phosphate cotransporter (NPT2A) mediates renal phosphate uptake. The NPT2A is regulated by parathyroid hormone (PTH) and fibroblast growth factor 23, which requires Na+/H+ exchange regulatory factor-1 (NHERF1), a multidomain PDZ-containing phosphoprotein. Phosphocycling controls the association between NHERF1 and the NPT2A. Here, we characterize the critical involvement of G protein-coupled receptor kinase 6A (GRK6A) in mediating PTH-sensitive phosphate transport by targeted phosphorylation coupled with NHERF1 conformational rearrangement, which in turn allows phosphorylation at a secondary site. GRK6A, through its carboxy-terminal PDZ recognition motif, binds NHERF1 PDZ1 with greater affinity than PDZ2. However, the association between NHERF1 PDZ2 and GRK6A is necessary for PTH action. Ser162, a PKCα phosphorylation site in PDZ2, regulates the binding affinity between PDZ2 and GRK6A. Substitution of Ser162 with alanine (S162A) blocks the PTH action but does not disrupt the interaction between NHERF1 and the NPT2A. Replacement of Ser162 with aspartic acid (S162D) abrogates the interaction between NHERF1 and the NPT2A and concurrently PTH action. We used amber codon suppression to generate a phosphorylated Ser162(pSer162)-PDZ2 variant. KD values determined by fluorescence anisotropy indicate that incorporation of pSer162 increased the binding affinity to the carboxy terminus of GRK6A 2-fold compared with WT PDZ2. Molecular dynamics simulations predict formation of an electrostatic network between pSer162 and Asp183 of PDZ2 and Arg at position -1 of the GRK6A PDZ-binding motif. Our results suggest that PDZ2 plays a regulatory role in PTH-sensitive NPT2A-mediated phosphate transport and phosphorylation of Ser162 in PDZ2 modulates the interaction with GRK6A.


Subject(s)
G-Protein-Coupled Receptor Kinases/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Biological Transport , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , G-Protein-Coupled Receptor Kinases/genetics , Humans , Ion Transport , Molecular Dynamics Simulation , PDZ Domains/genetics , Parathyroid Hormone/metabolism , Phosphates/metabolism , Phosphoproteins/genetics , Phosphorylation , Protein Binding , Protein Conformation , Sodium-Hydrogen Exchangers/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism
8.
Angew Chem Int Ed Engl ; 61(22): e202115805, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35299278

ABSTRACT

Sirtuin 5 (SIRT5) is a protein lysine deacylase enzyme that regulates diverse biology by hydrolyzing ϵ-N-carboxyacyllysine posttranslational modifications in the cell. Inhibition of SIRT5 has been linked to potential treatment of several cancers but potent compounds with activity in cells have been lacking. Here we developed mechanism-based inhibitors that incorporate isosteres of a carboxylic acid residue that is important for high-affinity binding to the enzyme active site. By masking of the tetrazole moiety of the most potent candidate from our initial SAR study, we achieved potent and cytoselective growth inhibition for the treatment of SIRT5-dependent leukemic cancer cell lines in culture. Thus, we provide an efficient, cellularly active small molecule that targets SIRT5, which can help elucidate its function and potential as a future drug target. This work shows that masked isosteres of carboxylic acids are viable chemical motifs for the development of inhibitors that target mitochondrial enzymes, which may have applications beyond the sirtuin field.


Subject(s)
Prodrugs , Sirtuins , Carboxylic Acids/pharmacology , Humans , Lysine/chemistry , Protein Processing, Post-Translational
9.
Angew Chem Int Ed Engl ; 61(47): e202204565, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36130196

ABSTRACT

The sirtuin enzymes are a family of lysine deacylases that regulate gene transcription and metabolism. Sirtuin 5 (SIRT5) hydrolyzes malonyl, succinyl, and glutaryl ϵ-N-carboxyacyllysine posttranslational modifications and has recently emerged as a vulnerability in certain cancers. However, chemical probes to illuminate its potential as a pharmacological target have been lacking. Here we report the harnessing of aryl fluorosulfate-based electrophiles as an avenue to furnish covalent inhibitors that target SIRT5. Alkyne-tagged affinity-labeling agents recognize and capture overexpressed SIRT5 in cultured HEK293T cells and can label SIRT5 in the hearts of mice upon intravenous injection of the compound. This work demonstrates the utility of aryl fluorosulfate electrophiles for targeting of SIRT5 and suggests this as a means for the development of potential covalent drug candidates. It is our hope that these results will serve as inspiration for future studies investigating SIRT5 and general sirtuin biology in the mitochondria.


Subject(s)
Neoplasms , Sirtuins , Humans , Animals , Mice , Lysine , HEK293 Cells , Sirtuins/chemistry , Neoplasms/genetics
10.
J Neurosci ; 40(25): 4954-4969, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32354853

ABSTRACT

Glycine receptors (GlyRs) are the major mediators of fast synaptic inhibition in the adult human spinal cord and brainstem. Hereditary mutations to GlyRs can lead to the rare, but potentially fatal, neuromotor disorder hyperekplexia. Most mutations located in the large intracellular domain (TM3-4 loop) of the GlyRα1 impair surface expression levels of the receptors. The novel GLRA1 mutation P366L, located in the TM3-4 loop, showed normal surface expression but reduced chloride currents, and accelerated whole-cell desensitization observed in whole-cell recordings. At the single-channel level, we observed reduced unitary conductance accompanied by spontaneous opening events in the absence of extracellular glycine. Using peptide microarrays and tandem MS-based analysis methods, we show that the proline-rich stretch surrounding P366 mediates binding to syndapin I, an F-BAR domain protein involved in membrane remodeling. The disruption of the noncanonical Src homology 3 recognition motif by P366L reduces syndapin I binding. These data suggest that the GlyRα1 subunit interacts with intracellular binding partners and may therefore play a role in receptor trafficking or synaptic anchoring, a function thus far only ascribed to the GlyRß subunit. Hence, the P366L GlyRα1 variant exhibits a unique set of properties that cumulatively affect GlyR functionality and thus might explain the neuropathological mechanism underlying hyperekplexia in the mutant carriers. P366L is the first dominant GLRA1 mutation identified within the GlyRα1 TM3-4 loop that affects GlyR physiology without altering protein expression at the whole-cell and surface levels.SIGNIFICANCE STATEMENT We show that the intracellular domain of the inhibitory glycine receptor α1 subunit contributes to trafficking and synaptic anchoring. A proline-rich stretch in this receptor domain forms a noncanonical recognition motif important for the interaction with syndapin I (PACSIN1). The disruption of this motif, as present in a human patient with hyperekplexia led to impaired syndapin I binding. Functional analysis revealed that the altered proline-rich stretch determines several functional physiological parameters of the ion channel (e.g., faster whole-cell desensitization) reduced unitary conductance and spontaneous opening events. Thus, the proline-rich stretch from the glycine receptor α1 subunit represents a multifunctional intracellular protein motif.


Subject(s)
Receptors, Glycine/genetics , Receptors, Glycine/metabolism , Stiff-Person Syndrome/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Animals , Humans , Mutation , Protein Binding/genetics , Protein Structure, Quaternary , Protein Transport/genetics , Receptors, Glycine/chemistry
11.
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
12.
Med Res Rev ; 40(1): 135-157, 2020 01.
Article in English | MEDLINE | ID: mdl-31218731

ABSTRACT

G proteins are key mediators of G protein-coupled receptor (GPCR) signaling, facilitating a plethora of important physiological processes. The role of G proteins is much less understood than other aspects of GPCR function, which is largely due to the shortage of potent and selective G protein inhibitors. The natural cyclic depsipeptides YM-254890 and FR900359 are two of the very few known selective inhibitors of the Gq subfamily, and are used as unique pharmacological tools in the study of G q -mediated signaling. Moreover, a peptide-based G protein antagonist-2A (GP-2A), a 27-residue peptide (27mer(I860A)) derived from phospholipase C-ß3 (PLC-ß3), and the small molecule BIM-46187 have also been characterized as selective G q inhibitors within the past 5 years. In this review, we highlight the recent development in chemical syntheses, characterization, and mechanism of action of these selective G q inhibitors. The development and application of G q -selective inhibitors will expand our knowledge of the structure and function of G protein-mediated signaling, shed light on the development of inhibitors for other G protein classes, and feed in to drug discovery for diseases where G proteins are implicated, including various forms of cancer.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Animals , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Proteins/metabolism , Humans , Models, Molecular , Structure-Activity Relationship
13.
J Biol Chem ; 294(15): 5747-5758, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30745359

ABSTRACT

Transmembrane signals initiated by a range of extracellular stimuli converge on members of the Gq family of heterotrimeric G proteins, which relay these signals in target cells. Gq family G proteins comprise Gq, G11, G14, and G16, which upon activation mediate their cellular effects via inositol lipid-dependent and -independent signaling to control fundamental processes in mammalian physiology. To date, highly specific inhibition of Gq/11/14 signaling can be achieved only with FR900359 (FR) and YM-254890 (YM), two naturally occurring cyclic depsipeptides. To further development of FR or YM mimics for other Gα subunits, we here set out to rationally design Gα16 proteins with artificial FR/YM sensitivity by introducing an engineered depsipeptide-binding site. Thereby we permit control of G16 function through ligands that are inactive on the WT protein. Using CRISPR/Cas9-generated Gαq/Gα11-null cells and loss- and gain-of-function mutagenesis along with label-free whole-cell biosensing, we determined the molecular coordinates for FR/YM inhibition of Gq and transplanted these to FR/YM-insensitive G16. Intriguingly, despite having close structural similarity, FR and YM yielded biologically distinct activities: it was more difficult to perturb Gq inhibition by FR and easier to install FR inhibition onto G16 than perturb or install inhibition with YM. A unique hydrophobic network utilized by FR accounted for these unexpected discrepancies. Our results suggest that non-Gq/11/14 proteins should be amenable to inhibition by FR scaffold-based inhibitors, provided that these inhibitors mimic the interaction of FR with Gα proteins harboring engineered FR-binding sites.


Subject(s)
Depsipeptides/pharmacology , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits , Peptides, Cyclic/pharmacology , Protein Engineering , Animals , CRISPR-Cas Systems , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Mice
14.
Nat Chem Biol ; 13(2): 153-160, 2017 02.
Article in English | MEDLINE | ID: mdl-27893705

ABSTRACT

γ-Aminobutyric acid type A and glycine receptors are the major mediators of fast synaptic inhibition in the human central nervous system and are established drug targets. However, all drugs targeting these receptors bind to the extracellular ligand-binding domain of the receptors, which inherently is associated with perturbation of the basic physiological action. Here we pursue a fundamentally different approach, by instead targeting the intracellular receptor-gephyrin interaction. First, we defined the gephyrin peptide-binding consensus sequence, which facilitated the development of gephyrin super-binding peptides and later effective affinity probes for the isolation of native gephyrin. Next, we demonstrated that fluorescent super-binding peptides could be used to directly visualize inhibitory postsynaptic sites for the first time in conventional and super-resolution microscopy. Finally, we demonstrate that the gephyrin super-binding peptides act as acute intracellular modulators of fast synaptic inhibition by modulating receptor clustering, thus being conceptually novel modulators of inhibitory neurotransmission.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Peptides/analysis , Peptides/pharmacology , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects , Animals , Carrier Proteins/chemistry , Cells, Cultured , HEK293 Cells , Humans , Luminescence , Membrane Proteins/chemistry , Mice , Peptides/chemical synthesis , Peptides/metabolism
15.
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.

16.
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
17.
Eur J Neurosci ; 45(2): 238-248, 2017 01.
Article in English | MEDLINE | ID: mdl-27859797

ABSTRACT

PSD-95 inhibitors have been shown to be neuroprotective in stroke, but have only to a very limited extent been evaluated in the treatment of traumatic brain injury (TBI) that has pathophysiological mechanisms in common with stroke. The aims of the current study were to assess the effects of a novel dimeric inhibitor of PSD-95, UCCB01-147, on histopathology and long-term cognitive outcome after controlled cortical impact (CCI) in rats. As excitotoxic cell death is thought to be a prominent part of the pathophysiology of TBI, we also investigated the neuroprotective effects of UCCB01-147 and related compounds on NMDA-induced cell death in cultured cortical neurons. Anesthetized rats were given a CCI or sham injury, and were randomized to receive an injection of either UCCB01-147 (10 mg/kg), the non-competitive NMDAR-receptor antagonist MK-801 (1 mg/kg) or saline immediately after injury. At 2 and 4 weeks post-trauma, spatial learning and memory were assessed in a water maze, and at 3 months, brains were removed for estimation of lesion volumes. Overall, neither treatment with UCCB01-147 nor MK-801 resulted in significant improvements of cognition and histopathology after CCI. Although MK-801 provided robust neuroprotection against NMDA-induced toxicity in cultured cortical neurons, UCCB01-147 failed to reduce cell death and became neurotoxic at high doses. The data suggest potential differential effects of PSD-95 inhibition in stroke and TBI that should be investigated further in future studies taking important experimental factors such as timing of treatment, dosage, and anesthesia into consideration.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Disks Large Homolog 4 Protein/antagonists & inhibitors , Dizocilpine Maleate/pharmacology , Motor Activity/drug effects , Neurons/drug effects , Animals , Brain/drug effects , Brain/physiopathology , Cognition/physiology , Disease Models, Animal , Male , Memory/drug effects , Motor Activity/physiology , Neuroprotective Agents/pharmacology , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/therapeutic use , Recovery of Function/drug effects
18.
Neurochem Res ; 42(12): 3401-3413, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28828633

ABSTRACT

Therapeutic effects of PSD-95 inhibition have been demonstrated in numerous studies of stroke; however only few studies have assessed the effects of PSD-95 inhibitors in traumatic brain injury (TBI). As the pathophysiology of TBI partially overlaps with that of stroke, PSD-95 inhibition may also be an effective therapeutic strategy in TBI. The objectives of the present study were to assess the effects of a dimeric inhibitor of PSD-95, UCCB01-144, on excitotoxic cell death in vitro and outcome after experimental TBI in rats in vivo. In addition, the pharmacokinetic parameters of UCCB01-144 were investigated in order to assess uptake of the drug into the central nervous system of rats. After a controlled cortical impact rats were randomized to receive a single injection of either saline or two different doses of UCCB01-144 (10 or 20 mg/kg IV) immediately after injury. Spatial learning and memory were assessed in a water maze at 2 weeks post-trauma, and at 4 weeks lesion volumes were estimated. Overall, UCCB01-144 did not protect against NMDA-toxicity in neuronal cultures or experimental TBI in rats. Important factors that should be investigated further in future studies assessing the effects of PSD-95 inhibitors in TBI are discussed.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Cell Death/drug effects , Disks Large Homolog 4 Protein/antagonists & inhibitors , Memory/drug effects , Oligopeptides/pharmacology , Animals , Disease Models, Animal , Maze Learning/drug effects , Mice , Neurons/drug effects
19.
J Biol Chem ; 290(23): 14582-94, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25903124

ABSTRACT

The serotonin transporter (SERT) terminates serotonergic neurotransmission by performing reuptake of released serotonin, and SERT is the primary target for antidepressants. SERT mediates the reuptake of serotonin through an alternating access mechanism, implying that a central substrate site is connected to both sides of the membrane by permeation pathways, of which only one is accessible at a time. The coordinated conformational changes in SERT associated with substrate translocation are not fully understood. Here, we have identified a Leu to Glu mutation at position 406 (L406E) in the extracellular loop 4 (EL4) of human SERT, which induced a remarkable gain-of-potency (up to >40-fold) for a range of SERT inhibitors. The effects were highly specific for L406E relative to six other mutations in the same position, including the closely related L406D mutation, showing that the effects induced by L406E are not simply charge-related effects. Leu(406) is located >10 Å from the central inhibitor binding site indicating that the mutation affects inhibitor binding in an indirect manner. We found that L406E decreased accessibility to a residue in the cytoplasmic pathway. The shift in equilibrium to favor a more outward-facing conformation of SERT can explain the reduced turnover rate and increased association rate of inhibitor binding we found for L406E. Together, our findings show that EL4 allosterically can modulate inhibitor binding within the central binding site, and substantiates that EL4 has an important role in controlling the conformational equilibrium of human SERT.


Subject(s)
Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Point Mutation , Protein Binding , Protein Conformation , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/pharmacology
20.
Chembiochem ; 17(20): 1936-1944, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27472651

ABSTRACT

PDZ domains are ubiquitous small protein domains that are mediators of numerous protein-protein interactions, and play a pivotal role in protein trafficking, synaptic transmission, and the assembly of signaling-transduction complexes. In recent years, PDZ domains have emerged as novel and exciting drug targets for diseases (in the brain in particular), so understanding the molecular details of PDZ domain interactions is of fundamental importance. PDZ domains bind to a protein partner at either a C-terminal peptide or internal peptide motifs. Here, we examined the importance of a conserved Lys/Arg residue in the ligand-binding site of the second PDZ domain of PSD-95, by employing a semisynthetic approach. We generated six semisynthetic PDZ domains comprising different proteogenic and nonproteogenic amino acids representing subtle changes of the conserved Lys/Arg residue. These were tested with four peptide interaction partners, representing the two different binding modes. The results highlight the role of a positively charged amino acid in the ß1-ß2 loop of PDZ domains, and show subtle differences for canonical and noncanonical interaction partners, thus providing additional insight into the mechanism of PDZ/ligand interaction.


Subject(s)
Dipeptides/metabolism , Membrane Proteins/biosynthesis , PDZ Domains , Binding Sites/drug effects , Dipeptides/chemistry , Humans , Ligands , Membrane Proteins/chemistry , Models, Molecular , PDZ Domains/drug effects , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL