Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 777
Filter
1.
Methods Enzymol ; 698: 247-262, 2024.
Article in English | MEDLINE | ID: mdl-38886034

ABSTRACT

The modulation of biology utilizing foldamers has flourished over the last few decades thanks to their overwhelming promise in their applications in molecular design, catalysis, supramolecular, and rational design. However, the application of peptidomimetics is still restricted due to the limited availability of molecular frameworks and folding propensities. To broaden the scope of foldameric peptidomimetics we proposed the development of sulfonyl-γ-AApeptides-the oligomers of sulfonyl-γ-N-acylated-N-aminoethyl (AA) amino acids, a unique unnatural scaffold that possesses promising potential to modulate protein-protein interactions. In this chapter, the overall process of design, synthesis, and function of sulfonyl-γ-AApeptides is briefly reviewed for the use of unnatural foldamers to modulate PPIs.


Subject(s)
Peptides , Peptidomimetics , Peptidomimetics/pharmacology , Peptidomimetics/chemistry , Peptides/chemistry , Peptides/pharmacology , Humans , Protein Folding , Amino Acids/chemistry , Amino Acids/metabolism , Protein Binding
2.
Nat Commun ; 15(1): 5275, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902227

ABSTRACT

DNA binding transcription factors possess the ability to interact with lipid membranes to construct ion-permeable pathways. Herein, we present a thiazole-based DNA binding peptide mimic TBP2, which forms transmembrane ion channels, impacting cellular ion concentration and consequently stabilizing G-quadruplex DNA structures. TBP2 self-assembles into nanostructures, e.g., vesicles and nanofibers and facilitates the transportation of Na+ and K+ across lipid membranes with high conductance (~0.6 nS). Moreover, TBP2 exhibits increased fluorescence when incorporated into the membrane or in cellular nuclei. Monomeric TBP2 can enter the lipid membrane and localize to the nuclei of cancer cells. The coordinated process of time-dependent membrane or nuclear localization of TBP2, combined with elevated intracellular cation levels and direct G-quadruplex (G4) interaction, synergistically promotes formation and stability of G4 structures, triggering cancer cell death. This study introduces a platform to mimic and control intricate biological functions, leading to the discovery of innovative therapeutic approaches.


Subject(s)
DNA , G-Quadruplexes , Peptidomimetics , Humans , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Peptidomimetics/metabolism , DNA/metabolism , DNA/chemistry , Potassium/metabolism , Potassium/chemistry , Cell Line, Tumor , Sodium/metabolism , Cell Nucleus/metabolism , Ion Channels/metabolism , Ion Channels/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry
3.
Molecules ; 29(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38893366

ABSTRACT

The development of antimicrobial drugs with novel structures and clear mechanisms of action that are active against drug-resistant bacteria has become an urgent need of safeguarding human health due to the rise of bacterial drug resistance. The discovery of AMPs and the development of amphipathic peptidomimetics have lay the foundation for novel antimicrobial agents to combat drug resistance due to their overall strong antimicrobial activities and unique membrane-active mechanisms. To break the limitation of AMPs, researchers have invested in great endeavors through various approaches in the past years. This review summarized the recent advances including the development of antibacterial small molecule peptidomimetics and peptide-mimic cationic oligomers/polymers, as well as mechanism-of-action studies. As this exciting interdisciplinary field is continuously expanding and growing, we hope this review will benefit researchers in the rational design of novel antimicrobial peptidomimetics in the future.


Subject(s)
Peptidomimetics , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Peptidomimetics/chemical synthesis , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Drug Resistance, Bacterial/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Microbial Sensitivity Tests , Bacteria/drug effects
4.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731881

ABSTRACT

Aging and age-related diseases are associated with a decline in the capacity of protein turnover. Intrinsically disordered proteins, as well as proteins misfolded and oxidatively damaged, prone to aggregation, are preferentially digested by the ubiquitin-independent proteasome system (UIPS), a major component of which is the 20S proteasome. Therefore, boosting 20S activity constitutes a promising strategy to counteract a decrease in total proteasome activity during aging. One way to enhance the proteolytic removal of unwanted proteins appears to be the use of peptide-based activators of the 20S. In this study, we synthesized a series of peptides and peptidomimetics based on the C-terminus of the Rpt5 subunit of the 19S regulatory particle. Some of them efficiently stimulated human 20S proteasome activity. The attachment of the cell-penetrating peptide TAT allowed them to penetrate the cell membrane and stimulate proteasome activity in HEK293T cells, which was demonstrated using a cell-permeable substrate of the proteasome, TAS3. Furthermore, the best activator enhanced the degradation of aggregation-prone α-synuclein and Tau-441. The obtained compounds may therefore have the potential to compensate for the unbalanced proteostasis found in aging and age-related diseases.


Subject(s)
Aging , Peptide Fragments , Proteasome Endopeptidase Complex , Humans , Aging/metabolism , alpha-Synuclein/metabolism , HEK293 Cells , Peptides/pharmacology , Peptides/chemistry , Peptides/metabolism , Peptidomimetics/pharmacology , Peptidomimetics/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Aggregates/drug effects , Protein Aggregation, Pathological/metabolism , Proteolysis/drug effects , tau Proteins/metabolism , Peptide Fragments/pharmacology
5.
Chemistry ; 30(38): e202401103, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38716707

ABSTRACT

This review covers the most recent advances in the development of inhibitors for the bacterial enzyme sortase A (SrtA). Sortase A (SrtA) is a critical virulence factor, present ubiquitously in Gram-positive bacteria of which many are pathogenic. Sortases are key enzymes regulating bacterial adherence to host cells, by anchoring extracellular matrix-binding proteins to the bacterial outer cell wall. By targeting virulence factors, effective treatment can be achieved, without inducing antibiotic resistance to the treatment. This is a potentially more sustainable, long-term approach to treating bacterial infections, including ones that display multiple resistance to current therapeutics. There are many promising approaches available for SrtA inhibition, some of which have the potential to advance into further clinical development, with peptidomimetic and in vivo active small molecules being among the most promising. There are currently no approved drugs on the market targeting SrtA, despite its promise, adding to the relevance of this review article, as it extends to the pharmaceutical industry additionally to academic researchers.


Subject(s)
Aminoacyltransferases , Anti-Bacterial Agents , Bacterial Proteins , Cysteine Endopeptidases , Peptidomimetics , Small Molecule Libraries , Aminoacyltransferases/antagonists & inhibitors , Aminoacyltransferases/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/chemistry , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Humans , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gram-Positive Bacteria/drug effects
6.
J Med Chem ; 67(11): 8757-8790, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38753594

ABSTRACT

Given the crucial role of the main protease (Mpro) in the replication cycle of SARS-CoV-2, this viral cysteine protease constitutes a high-profile drug target. We investigated peptidomimetic azapeptide nitriles as auspicious, irreversibly acting inhibitors of Mpro. Our systematic approach combined an Mpro active-site scanning by combinatorially assembled azanitriles with structure-based design. Encouraged by the bioactive conformation of open-chain inhibitors, we conceptualized the novel chemotype of macrocyclic azanitriles whose binding mode was elucidated by cocrystallization. This strategy provided a favorable entropic contribution to target binding and resulted in the development of the extraordinarily potent Mpro inhibitor 84 with an IC50 value of 3.23 nM and a second-order rate constant of inactivation, kinac/Ki, of 448,000 M-1s-1. The open-chain Mpro inhibitor 58, along with the macrocyclic compounds 83 and 84, a broad-spectrum anticoronaviral agent, demonstrated the highest antiviral activity with EC50 values in the single-digit micromolar range. Our findings are expected to promote the future development of peptidomimetic Mpro inhibitors as anti-SARS-CoV-2 agents.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases , Nitriles , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , SARS-CoV-2/drug effects , Nitriles/chemistry , Nitriles/pharmacology , Nitriles/chemical synthesis , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , Structure-Activity Relationship , Humans , Macrocyclic Compounds/pharmacology , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/chemical synthesis , COVID-19 Drug Treatment , Drug Discovery , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/chemical synthesis , Peptidomimetics/pharmacology , Peptidomimetics/chemistry , Peptidomimetics/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacology , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Peptides/chemical synthesis
7.
J Med Chem ; 67(11): 8585-8608, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38809766

ABSTRACT

The von Hippel-Lindau (VHL) protein plays a pivotal role in regulating the hypoxic stress response and has been extensively studied and utilized in the targeted protein degradation field, particularly in the context of bivalent degraders. In this study, we present a comprehensive peptidomimetic structure-activity relationship (SAR) approach, combined with cellular NanoBRET target engagement assays to enhance the existing VHL ligands. Through systematic modifications of the molecule, we identified the 1,2,3-triazole group as an optimal substitute of the left-hand side amide bond that yields 10-fold higher binding activity. Moreover, incorporating conformationally constrained alterations on the methylthiazole benzylamine moiety led to the development of highly potent VHL ligands with picomolar binding affinity and significantly improved oral bioavailability. We anticipate that our optimized VHL ligand, GNE7599, will serve as a valuable tool compound for investigating the VHL pathway and advancing the field of targeted protein degradation.


Subject(s)
Biological Availability , Peptidomimetics , Von Hippel-Lindau Tumor Suppressor Protein , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/chemistry , Peptidomimetics/chemistry , Peptidomimetics/pharmacokinetics , Peptidomimetics/pharmacology , Humans , Ligands , Structure-Activity Relationship , Administration, Oral , Animals
8.
J Agric Food Chem ; 72(20): 11341-11350, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38713071

ABSTRACT

Insect neuropeptides play an essential role in regulating growth, development, reproduction, nerve conduction, metabolism, and behavior in insects; therefore, G protein-coupled receptors of neuropeptides are considered important targets for designing green insecticides. Cockroach-type allatostatins (ASTs) (FGLamides allatostatins) are important insect neuropeptides in Diploptera punctata that inhibit juvenile hormone (JH) synthesis in the corpora allata and affect growth, development, and reproduction of insects. Therefore, the pursuit of novel insecticides targeting the allatostatin receptor (AstR) holds significant importance. Previously, we identified an AST analogue, H17, as a promising candidate for pest control. Herein, we first modeled the 3D structure of AstR in D. punctata (Dippu-AstR) and predicted the binding mode of H17 with Dippu-AstR to study the critical interactions and residues favorable to its bioactivity. Based on this binding mode, we designed and synthesized a series of H17 derivatives and assessed their insecticidal activity against D. punctata. Among them, compound Q6 showed higher insecticidal activity than H17 against D. punctata by inhibiting JH biosynthesis, indicating that Q6 is a potential candidate for a novel insect growth regulator (IGR)-based insecticide. Moreover, Q6 exhibited insecticidal activity against Plutella xylostella, indicating that these AST analogs may have a wider insecticidal spectrum. The underlying mechanisms and molecular conformations mediating the interactions of Q6 with Dippu-AstR were explored to understand its effects on the bioactivity. The present work clarifies how a target-based strategy facilitates the discovery of new peptide mimics with better bioactivity, enabling improved IGR-based insecticide potency in sustainable agriculture.


Subject(s)
Insect Proteins , Insecticides , Neuropeptides , Peptidomimetics , Insecticides/chemistry , Insecticides/pharmacology , Insecticides/chemical synthesis , Animals , Neuropeptides/chemistry , Neuropeptides/pharmacology , Neuropeptides/metabolism , Insect Proteins/chemistry , Insect Proteins/metabolism , Insect Proteins/genetics , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Peptidomimetics/chemical synthesis , Drug Design , Juvenile Hormones/chemistry , Juvenile Hormones/pharmacology , Juvenile Hormones/metabolism , Cockroaches/drug effects , Cockroaches/chemistry
9.
Org Lett ; 26(19): 4088-4092, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38709636

ABSTRACT

Access to 1,2,3-triazolium-grafted peptoid macrocycles was developed by macrocyclization and multivalent postmodification of linear peptoid oligomers carrying an alternance of benzylic and propargyl groups as side chains. X-ray analysis and NMR studies revealed a conformational preference for constrained hairpin-shaped structures leading to the facial amphipathic character of these macrocycles. A preliminary evaluation showed the antimicrobial activities of these new cationic amphipathic architectures.


Subject(s)
Anti-Bacterial Agents , Macrocyclic Compounds , Microbial Sensitivity Tests , Peptidomimetics , Triazoles , Triazoles/chemistry , Triazoles/pharmacology , Molecular Structure , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Peptidomimetics/chemical synthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Macrocyclic Compounds/chemical synthesis , Peptoids/chemistry , Peptoids/pharmacology , Peptoids/chemical synthesis , Crystallography, X-Ray , Bacteria/drug effects
10.
Elife ; 122024 Apr 29.
Article in English | MEDLINE | ID: mdl-38682900

ABSTRACT

The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the 'WIN' site that tethers WDR5 to chromatin. WIN site inhibitors (WINi) are active against multiple cancer cell types in vitro, the most notable of which are those derived from MLL-rearranged (MLLr) leukemias. Peptidomimetic WINi were originally proposed to inhibit MLLr cells via dysregulation of genes connected to hematopoietic stem cell expansion. Our discovery and interrogation of small-molecule WINi, however, revealed that they act in MLLr cell lines to suppress ribosome protein gene (RPG) transcription, induce nucleolar stress, and activate p53. Because there is no precedent for an anticancer strategy that specifically targets RPG expression, we took an integrated multi-omics approach to further interrogate the mechanism of action of WINi in human MLLr cancer cells. We show that WINi induce depletion of the stock of ribosomes, accompanied by a broad yet modest translational choke and changes in alternative mRNA splicing that inactivate the p53 antagonist MDM4. We also show that WINi are synergistic with agents including venetoclax and BET-bromodomain inhibitors. Together, these studies reinforce the concept that WINi are a novel type of ribosome-directed anticancer therapy and provide a resource to support their clinical implementation in MLLr leukemias and other malignancies.


Subject(s)
Intracellular Signaling Peptides and Proteins , Myeloid-Lymphoid Leukemia Protein , Nuclear Proteins , Ribosomes , Tumor Suppressor Protein p53 , Humans , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Peptidomimetics/pharmacology
11.
J Virol ; 98(5): e0190323, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38593045

ABSTRACT

We developed a novel class of peptidomimetic inhibitors targeting several host cell human serine proteases, including transmembrane protease serine 2 (TMPRSS2), matriptase, and hepsin. TMPRSS2 is a membrane-associated protease that is highly expressed in the upper and lower respiratory tracts and is utilized by SARS-CoV-2 and other viruses to proteolytically process their glycoproteins, enabling host cell entry, replication, and dissemination of new virus particles. We have previously shown that compound MM3122 exhibited subnanomolar potency against all three proteases and displayed potent antiviral effects against SARS-CoV-2 in a cell viability assay. Herein, we demonstrate that MM3122 potently inhibits viral replication in human lung epithelial cells and is also effective against the EG.5.1 variant of SARS-CoV-2. Furthermore, we evaluated MM3122 in a mouse model of COVID-19 and demonstrated that MM3122 administered intraperitoneally (IP) before (prophylactic) or after (therapeutic) SARS-CoV-2 infection had significant protective effects against weight loss and lung congestion and reduced pathology. Amelioration of COVID-19 disease was associated with a reduction in proinflammatory cytokine and chemokine production after SARS-CoV-2 infection. Prophylactic, but not therapeutic, administration of MM3122 also reduced virus titers in the lungs of SARS-CoV-2-infected mice. Therefore, MM3122 is a promising lead candidate small-molecule drug for the treatment and prevention of infections caused by SARS-CoV-2 and other coronaviruses. IMPORTANCE: SARS-CoV-2 and other emerging RNA coronaviruses are a present and future threat in causing widespread endemic and pandemic infection and disease. In this paper, we have shown that the novel host cell protease inhibitor, MM3122, blocks SARS-CoV-2 viral replication and is efficacious as both a prophylactic and a therapeutic drug for the treatment of COVID-19 given intraperitoneally in mice. Targeting host proteins and pathways in antiviral therapy is an underexplored area of research, but this approach promises to avoid drug resistance by the virus, which is common in current antiviral treatments.


Subject(s)
Antiviral Agents , Benzothiazoles , COVID-19 Drug Treatment , Oligopeptides , SARS-CoV-2 , Serine Proteinase Inhibitors , Virus Replication , Animals , Female , Humans , Mice , Antiviral Agents/pharmacology , Chlorocebus aethiops , COVID-19/virology , Disease Models, Animal , Lung/virology , Lung/pathology , Lung/drug effects , Peptidomimetics/pharmacology , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Vero Cells , Virus Replication/drug effects , Oligopeptides/pharmacology , Benzothiazoles/pharmacology
12.
Bioorg Chem ; 147: 107316, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38583246

ABSTRACT

Ras GTPases and other CaaX proteins undergo multiple post-translational modifications at their carboxyl-terminus. These events initiate with prenylation of a cysteine and are followed by endoproteolytic removal of the 'aaX' tripeptide and carboxylmethylation. Some CaaX proteins are only subject to prenylation, however, due to the presence of an uncleavable sequence. In this study, uncleavable sequences were used to stage Ras isoforms in a farnesylated and uncleaved state to address the impact of CaaX proteolysis on protein localization and function. This targeted strategy is more specific than those that chemically inhibit the Rce1 CaaX protease or delete the RCE1 gene because global abrogation of CaaX proteolysis impacts the entire CaaX protein proteome and effects cannot be attributed to any specific CaaX protein of the many concurrently affected. With this targeted strategy, clear mislocalization and reduced activity of farnesylated and uncleaved Ras isoforms was observed. In addition, new peptidomimetics based on cleavable Ras CaaX sequences and the uncleavable CAHQ sequence were synthesized and tested as Rce1 inhibitors using in vitro and cell-based assays. Consistently, these non-hydrolyzable peptidomimetic Rce1 inhibitors recapitulate Ras mislocalization effects when modeled on cleavable but not uncleavable CaaX sequences. These findings indicate that a prenylated and uncleavable CaaX sequence, which can be easily applied to a wide range of mammalian CaaX proteins, can be used to probe the specific impact of CaaX proteolysis on CaaX protein properties under conditions of an otherwise normally processed CaaX protein proteome.


Subject(s)
ras Proteins , Humans , ras Proteins/metabolism , ras Proteins/antagonists & inhibitors , ras Proteins/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Proteolysis/drug effects , Molecular Structure , Peptidomimetics/pharmacology , Peptidomimetics/chemistry , Peptidomimetics/chemical synthesis , Endopeptidases
13.
Sci Rep ; 14(1): 8991, 2024 04 18.
Article in English | MEDLINE | ID: mdl-38637583

ABSTRACT

COVID-19 is a multisystemic disease caused by the SARS-CoV-2 airborne virus, a member of the Coronaviridae family. It has a positive sense single-stranded RNA genome and encodes two non-structural proteins through viral cysteine-proteases processing. Blocking this step is crucial to control virus replication. In this work, we reported the synthesis of 23 statine-based peptidomimetics to determine their ability to inhibit the main protease (Mpro) activity of SARS-CoV-2. Among the 23 peptidomimetics, 15 compounds effectively inhibited Mpro activity by 50% or more, while three compounds (7d, 8e, and 9g) exhibited maximum inhibition above 70% and IC50 < 1 µM. Compounds 7d, 8e, and 9g inhibited roughly 80% of SARS-CoV-2 replication and proved no cytotoxicity. Molecular docking simulations show putative hydrogen bond and hydrophobic interactions between specific amino acids and these inhibitors. Molecular dynamics simulations further confirmed the stability and persisting interactions in Mpro's subsites, exhibiting favorable free energy binding (ΔGbind) values. These findings suggest the statine-based peptidomimetics as potential therapeutic agents against SARS-CoV-2 by targeting Mpro.


Subject(s)
COVID-19 , Coronavirus 3C Proteases , Peptidomimetics , Humans , SARS-CoV-2/metabolism , Peptidomimetics/pharmacology , Molecular Docking Simulation , Protease Inhibitors/chemistry , Amino Acids , Molecular Dynamics Simulation , Antiviral Agents/pharmacology , Antiviral Agents/chemistry
14.
Eur J Med Chem ; 268: 116263, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38432056

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and related variants, are responsible for the devastating coronavirus disease 2019 (COVID-19) pandemic. The SARS-CoV-2 main protease (Mpro) plays a central role in the replication of the virus and represents an attractive drug target. Herein, we report the discovery of novel SARS-CoV-2 Mpro covalent inhibitors, including highly effective compound NIP-22c which displays high potency against several key variants and clinically relevant nirmatrelvir Mpro E166V mutants.


Subject(s)
COVID-19 , Peptidomimetics , Humans , Peptidomimetics/pharmacology , Peptide Hydrolases , Protease Inhibitors/pharmacology , SARS-CoV-2 , Cysteine Endopeptidases , Antiviral Agents/pharmacology
15.
Int J Mol Sci ; 25(3)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38338655

ABSTRACT

Trypsin-like serine proteases are involved in many important physiological processes like blood coagulation and remodeling of the extracellular matrix. On the other hand, they are also associated with pathological conditions. The urokinase-pwlasminogen activator (uPA), which is involved in tissue remodeling, can increase the metastatic behavior of various cancer types when overexpressed and dysregulated. Another member of this protease class that received attention during the SARS-CoV 2 pandemic is TMPRSS2. It is a transmembrane serine protease, which enables cell entry of the coronavirus by processing its spike protein. A variety of different inhibitors have been published against both proteases. However, the selectivity over other trypsin-like serine proteases remains a major challenge. In the current study, we replaced the arginine moiety at the P1 site of peptidomimetic inhibitors with different bioisosteres. Enzyme inhibition studies revealed that the phenylguanidine moiety in the P1 site led to strong affinity for TMPRSS2, whereas the cyclohexylguanidine derivate potently inhibited uPA. Both inhibitors exhibited high selectivity over other structurally similar and physiologically important proteases.


Subject(s)
Peptidomimetics , Serine Proteinase Inhibitors , Urokinase-Type Plasminogen Activator , Ligands , Peptide Hydrolases , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Trypsin , Urokinase-Type Plasminogen Activator/metabolism , Serine Endopeptidases , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology
16.
Int J Mol Sci ; 25(2)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38256156

ABSTRACT

Peptides are increasingly emerging as a drug class for a wide range of human diseases due to their intrinsic properties, such as excellent recognition abilities and biocompatibility [...].


Subject(s)
Peptidomimetics , Humans , Peptidomimetics/pharmacology , Research , Peptides , Recognition, Psychology
17.
Sci Rep ; 14(1): 2572, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38296985

ABSTRACT

Bacterial caseinolytic protease P subunit (ClpP) is important and vital for cell survival and infectivity. Recent publications describe and discuss the complex structure-function relationship of ClpP and its processive activity mediated by 14 catalytic sites. Even so, there are several aspects yet to be further elucidated, such as the paradoxical allosteric modulation of ClpP by peptidomimetic boronates. These compounds bind to all catalytic sites, and in specific conditions, they stimulate a dysregulated degradation of peptides and globular proteins, instead of inhibiting the enzymatic activity, as expected for serine proteases in general. Aiming to explore and explain this paradoxical effect, we solved and refined the crystal structure of native ClpP from Staphylococcus epidermidis (Se), an opportunistic pathogen involved in nosocomial infections, as well as ClpP in complex with ixazomib at 1.90 Å and 2.33 Å resolution, respectively. The interpretation of the crystal structures, in combination with complementary biochemical and biophysical data, shed light on how ixazomib affects the ClpP conformational state and activity. Moreover, SEC-SAXS and DLS measurements show, for the first time, that a peptidomimetic boronate compound also induces the assembly of the tetradecameric structure from isolated homomeric heptameric rings of a gram-positive organism.


Subject(s)
Glycine/analogs & derivatives , Peptidomimetics , Peptidomimetics/pharmacology , Scattering, Small Angle , X-Ray Diffraction , Boron Compounds/pharmacology , Boron Compounds/metabolism , Endopeptidase Clp/metabolism , Bacterial Proteins/metabolism
18.
Eur J Med Chem ; 266: 116128, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38232463

ABSTRACT

In this paper we present the design, synthesis, and biological evaluation of a new series of peptidomimetics acting as potent anti-SARS-CoV-2 agents. Starting from our previously described Main Protease (MPro) and Papain Like Protease (PLPro) dual inhibitor, CV11, here we disclose its high inhibitory activity against cathepsin L (CTSL) (IC50 = 19.80 ± 4.44 nM), an emerging target in SARS-CoV-2 infection machinery. An in silico design, inspired by the structure of CV11, led to the development of a library of peptidomimetics showing interesting activities against CTSL and Mpro, allowing us to trace the chemical requirements for the binding to both enzymes. The screening in Vero cells infected with 5 different SARS-CoV-2 variants of concerns, highlighted sub-micromolar activities for most of the synthesized compounds (13, 15, 16, 17 and 31) in agreement with the enzymatic inhibition assays results. The compounds showed lack of activity against several different RNA viruses except for the 229E and OC43 human coronavirus strains, also characterized by a cathepsin-L dependent release into the host cells. The most promising derivatives were also evaluated for their chemical and metabolic in-vitro stability, with derivatives 15 and 17 showing a suitable profile for further preclinical characterization.


Subject(s)
COVID-19 , Peptidomimetics , Chlorocebus aethiops , Humans , Animals , Cathepsin L , SARS-CoV-2 , Peptidomimetics/pharmacology , Protease Inhibitors/pharmacology , Vero Cells , Peptide Hydrolases , Antiviral Agents/pharmacology , Molecular Docking Simulation
19.
J Med Chem ; 67(2): 885-921, 2024 01 25.
Article in English | MEDLINE | ID: mdl-38179950

ABSTRACT

Hepatitis C viral (HCV) infection is the leading cause of liver failure and still represents a global health burden. Over the past decade, great advancements made HCV curable, and sustained viral remission significantly improved to more than 98%. Historical treatment with pegylated interferon alpha and ribavirin has been displaced by combinations of direct-acting antivirals. These regimens include drugs targeting different stages of the HCV life cycle. However, the emergence of viral resistance remains a big concern. The design of peptidomimetic inhibitors (PIs) able to fit and fill the conserved substrate envelope region within the active site helped avoid contact with the vulnerable sites of the most common resistance-associated substitutions Arg155, Ala156, and Asp168. Herein, we give an overview of HCV NS3 PIs discovered during the past decade, and we deeply discuss the rationale behind the structural optimization efforts essential to achieve pangenotypic activity.


Subject(s)
Hepatitis C, Chronic , Hepatitis C , Peptidomimetics , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Antiviral Agents/chemistry , Peptidomimetics/pharmacology , Peptidomimetics/therapeutic use , Protease Inhibitors/pharmacology , Hepatitis C, Chronic/drug therapy , Viral Nonstructural Proteins , Hepatitis C/drug therapy , Hepacivirus , Drug Resistance, Viral
20.
Arch Pharm (Weinheim) ; 357(4): e2300503, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38251950

ABSTRACT

A series of 1,1'-biphenyl-3-carboxamide and furan-phenyl-carboxamide analogs were synthesized using an optimized scheme and confirmed by 1H and 13C nuclear magnetic resonance and high-resolution mass spectrometry techniques. The synthesized peptidomimetics analogs were screened in vitro to understand the inhibitory potential of pancreatic lipase (PL). Analogs were assessed for the PL inhibitory activity based on interactions, geometric complementarity, and docking score. Among the synthesized analogs, 9, 29, and 24 were found to have the most potent PL inhibitory activity with IC50 values of 3.87, 4.95, and 5.34 µM, respectively, compared to that of the standard drug, that is, orlistat, which inhibits PL with an IC50 value of 0.99 µM. The most potent analog, 9, exhibited a competitive-type inhibition with an inhibition constant (Ki) of 2.72 µM. In silico molecular docking of analog 9 with the PL (PDB ID:1LPB) showed a docking score of -11.00 kcal/mol. Analog 9 formed crucial hydrogen bond interaction with Ser152, His263, π-cation interaction with Asp79, Arg256, and π-π stacking with Phe77, Tyr114 at the protein's active site. The molecular dynamic simulation confirmed that analog 9 forms stable interactions with PL at the end of 200 ns with root mean square deviation values of 2.5 and 6 Å. No toxicity was observed for analog 9 (concentration range of 1-20 µM) when tested by MTT assay in RAW 264.7 cells.


Subject(s)
Peptidomimetics , Humans , Structure-Activity Relationship , Peptidomimetics/pharmacology , Molecular Docking Simulation , Lipase , Obesity/drug therapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...