Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Chem Zvesti ; 77(3): 1695-1702, 2023.
Article in English | MEDLINE | ID: mdl-36466109

ABSTRACT

Favipiravir is a wide-spectrum antiviral generic drug that has received large attention during the recent COVID-19 pandemic. While there are synthetic strategies for favipiravir synthesis, economical procedures could contribute to industrial scale synthesis and availability. Accordingly, our efforts focused on an economic and scalable procedure for favipiravir synthesis via the 3,6-dichloropyrazine-2-carbonitrile intermediate obtained from 3-aminopyrazine-2-carboxylic acid. The process afforded favipiravir with 43% yield (from 3,6-dichloropyrazine-2-carbonitrile, by fluorination, hydroxylation, and nitrile hydrolysis reactions) and greater than 99% purity without a chromatographic purification step. Supplementary Information: The online version contains supplementary material available at 10.1007/s11696-022-02595-1.

2.
Angew Chem Int Ed Engl ; 61(18): e202115193, 2022 04 25.
Article in English | MEDLINE | ID: mdl-35170181

ABSTRACT

For the discovery of novel chemical matter generally endowed with bioactivity, strategies may be particularly efficient that combine previous insight about biological relevance, e.g., natural product (NP) structure, with methods that enable efficient coverage of chemical space, such as fragment-based design. We describe the de novo combination of different 5-membered NP-derived N-heteroatom fragments to structurally unprecedented "pseudo-natural products" in an efficient complexity-generating and enantioselective one-pot synthesis sequence. The pseudo-NPs inherit characteristic elements of NP structure but occupy areas of chemical space not covered by NP-derived chemotypes, and may have novel biological targets. Investigation of the pseudo-NPs in unbiased phenotypic assays and target identification led to the discovery of the first small-molecule ligand of the RHO GDP-dissociation inhibitor 1 (RHOGDI1), termed Rhonin. Rhonin inhibits the binding of the RHOGDI1 chaperone to GDP-bound RHO GTPases and alters the subcellular localization of RHO GTPases.


Subject(s)
Biological Products , Biological Products/chemistry , Ligands , rho GTP-Binding Proteins , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Specific Guanine Nucleotide Dissociation Inhibitors
3.
Nat Commun ; 11(1): 5425, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110077

ABSTRACT

Transcription factors are key protein effectors in the regulation of gene transcription, and in many cases their activity is regulated via a complex network of protein-protein interactions (PPI). The chemical modulation of transcription factor activity is a long-standing goal in drug discovery but hampered by the difficulties associated with the targeting of PPIs, in particular when extended and flat protein interfaces are involved. Peptidomimetics have been applied to inhibit PPIs, however with variable success, as for certain interfaces the mimicry of a single secondary structure element is insufficient to obtain high binding affinities. Here, we describe the design and characterization of a stabilized protein tertiary structure that acts as an inhibitor of the interaction between the transcription factor TEAD and its co-repressor VGL4, both playing a central role in the Hippo signalling pathway. Modification of the inhibitor with a cell-penetrating entity yielded a cell-permeable proteomimetic that activates cell proliferation via regulation of the Hippo pathway, highlighting the potential of protein tertiary structure mimetics as an emerging class of PPI modulators.


Subject(s)
Peptidomimetics , Transcription Factors/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Hippo Signaling Pathway , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
4.
J Med Chem ; 63(20): 11972-11989, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32907324

ABSTRACT

Transcriptional enhanced associate domain (TEAD) transcription factors together with coactivators and corepressors modulate the expression of genes that regulate fundamental processes, such as organogenesis and cell growth, and elevated TEAD activity is associated with tumorigenesis. Hence, novel modulators of TEAD and methods for their identification are in high demand. We describe the development of a new "thiol conjugation assay" for identification of novel small molecules that bind to the TEAD central pocket. The assay monitors prevention of covalent binding of a fluorescence turn-on probe to a cysteine in the central pocket by small molecules. Screening of a collection of compounds revealed kojic acid analogues as TEAD inhibitors, which covalently target the cysteine in the central pocket, block the interaction with coactivator yes-associated protein with nanomolar apparent IC50 values, and reduce TEAD target gene expression. This methodology promises to enable new medicinal chemistry programs aimed at the modulation of TEAD activity.


Subject(s)
Drug Discovery , Pyrones/pharmacology , Small Molecule Libraries/pharmacology , Sulfhydryl Compounds/pharmacology , Transcription Factors/antagonists & inhibitors , Dose-Response Relationship, Drug , Fluorescence , Humans , Models, Molecular , Molecular Structure , Pyrones/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , Transcription Factors/genetics
5.
Angew Chem Int Ed Engl ; 59(46): 20338-20342, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32537835

ABSTRACT

DNA-encoded combinatorial synthesis provides efficient and dense coverage of chemical space around privileged molecular structures. The indole side chain of tryptophan plays a prominent role in key, or "hot spot", regions of protein-protein interactions. A DNA-encoded combinatorial peptoid library was designed based on the Ugi four-component reaction by employing tryptophan-mimetic indole side chains to probe the surface of target proteins. Several peptoids were synthesized on a chemically stable hexathymidine adapter oligonucleotide "hexT", encoded by DNA sequences, and substituted by azide-alkyne cycloaddition to yield a library of 8112 molecules. Selection experiments for the tumor-relevant proteins MDM2 and TEAD4 yielded MDM2 binders and a novel class of TEAD-YAP interaction inhibitors that perturbed the expression of a gene under the control of these Hippo pathway effectors.


Subject(s)
DNA/metabolism , Indoles/metabolism , Peptidomimetics , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription Factors/metabolism , Humans , Protein Binding
6.
ACS Chem Biol ; 14(12): 2909-2921, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31742995

ABSTRACT

The transcriptional co-regulators YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif) are the vertebrate downstream effectors of the Hippo signaling pathway that controls various physiological and pathological processes. YAP and TAZ pair with the TEAD (TEA domain) family of transcription factors to initiate transcription. We previously identified a tractable pocket in TEADs, which has been physiologically shown to bind palmitate. Herein, a TEAD-palmitate interaction screen was developed to select small molecules occupying the palmitate-binding pocket (PBP) of TEADs. We show that quinolinols were TEAD-binding compounds that augment YAP/TAZ-TEAD activity, which was verified using TEAD reporter assay, RT-qPCR, and RNA-Seq analyses. Structure-activity relationship investigations uncovered the quinolinol substituents that are necessary for TEAD activation. We reveal a novel mechanism where quinolinols stabilize YAP/TAZ protein levels by occupying the PBP. The enhancement of YAP activity by quinolinols accelerates the in vivo wound closure in a mouse wound-healing model. Although small molecules that occupy the PBP have been shown to inhibit YAP/TAZ-TEAD activity, leveraging PBP to activate TEADs is a novel approach.


Subject(s)
Hydroxyquinolines/pharmacology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Animals , HEK293 Cells , Humans , Hydroxyquinolines/chemistry , Mice , Mice, Inbred ICR , Skin/drug effects , Structure-Activity Relationship , Wound Healing/drug effects
7.
ACS Chem Biol ; 14(10): 2197-2205, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31498986

ABSTRACT

Peptides constitute an important class of drugs for the treatment of multiple metabolic, oncological, and neurodegenerative diseases, and several hundred novel therapeutic peptides are currently in the preclinical and clinical stages of development. However, many leads fail to advance clinically because of poor cellular membrane and tissue permeability. Therefore, assessment of the ability of a peptide to cross cellular membranes is critical when developing novel peptide-based therapeutics. Current methods to assess peptide cellular permeability are limited by multiple factors, such as the need to introduce rather large modifications (e.g., fluorescent dyes) that require complex chemical reactions as well as an inability to provide kinetic information on the internalization of a compound or distinguish between internalized and membrane-bound compounds. In addition, many of these methods are based on end point assays and require multiple sample manipulation steps. Herein, we report a novel "Split Luciferin Peptide" (SLP) assay that enables the real-time noninvasive imaging and quantification of peptide uptake both in vitro and in vivo using a very sensitive bioluminescence readout. This method is based on a straightforward, stable chemical modification of the peptide of interest with a d-cysteine tag that preserves the overall peptidic character of the original molecule. This method can be easily adapted for screening peptide libraries and can thus become an important tool for preclinical peptide drug development.


Subject(s)
Biological Assay/methods , Luminescent Measurements/methods , Peptides/analysis , Animals , Cell Line, Tumor , Cysteine/chemistry , Female , Firefly Luciferin/metabolism , Humans , Luciferases, Firefly/metabolism , Mice , Nitriles/chemistry , Peptides/chemistry , Peptides/metabolism , Protein Transport
8.
Nat Chem Biol ; 15(7): 710-720, 2019 07.
Article in English | MEDLINE | ID: mdl-31222192

ABSTRACT

Autophagy mediates the degradation of damaged proteins, organelles and pathogens, and plays a key role in health and disease. Thus, the identification of new mechanisms involved in the regulation of autophagy is of major interest. In particular, little is known about the role of lipids and lipid-binding proteins in the early steps of autophagosome biogenesis. Using target-agnostic, high-content, image-based identification of indicative phenotypic changes induced by small molecules, we have identified autogramins as a new class of autophagy inhibitor. Autogramins selectively target the recently discovered cholesterol transfer protein GRAM domain-containing protein 1A (GRAMD1A, which had not previously been implicated in autophagy), and directly compete with cholesterol binding to the GRAMD1A StART domain. GRAMD1A accumulates at sites of autophagosome initiation, affects cholesterol distribution in response to starvation and is required for autophagosome biogenesis. These findings identify a new biological function of GRAMD1A and a new role for cholesterol in autophagy.


Subject(s)
Autophagosomes/metabolism , Membrane Proteins/metabolism , Autophagosomes/drug effects , Autophagy/drug effects , Humans , Membrane Proteins/antagonists & inhibitors , Models, Molecular , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured
9.
J Med Chem ; 60(12): 4818-4839, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28603984

ABSTRACT

We report herein the design, synthesis, and evaluation of macrocyclic peptidomimetics that bind to WD repeat domain 5 (WDR5) and block the WDR5-mixed lineage leukemia (MLL) protein-protein interaction. Compound 18 (MM-589) binds to WDR5 with an IC50 value of 0.90 nM (Ki value <1 nM) and inhibits the MLL H3K4 methyltransferase (HMT) activity with an IC50 value of 12.7 nM. Compound 18 potently and selectively inhibits cell growth in human leukemia cell lines harboring MLL translocations and is >40 times better than the previously reported compound MM-401. Cocrystal structures of 16 and 18 complexed with WDR5 provide structural basis for their high affinity binding to WDR5. Additionally, we have developed and optimized a new AlphaLISA-based MLL HMT functional assay to facilitate the functional evaluation of these designed compounds. Compound 18 represents the most potent inhibitor of the WDR5-MLL interaction reported to date, and further optimization of 18 may yield a new therapy for acute leukemia.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Peptides, Cyclic/pharmacology , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Animals , Binding, Competitive , Cell Line, Tumor , Cell Survival/drug effects , Chemistry Techniques, Synthetic , Drug Discovery , Drug Stability , High-Throughput Screening Assays/methods , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Intracellular Signaling Peptides and Proteins , Leukemia/drug therapy , Leukemia/pathology , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Magnetic Resonance Spectroscopy , Mice , Microsomes/drug effects , Molecular Docking Simulation , Myeloid-Lymphoid Leukemia Protein/genetics , Peptides, Cyclic/chemistry , Peptidomimetics/metabolism , Protein Interaction Domains and Motifs , Rats
10.
Trends Biotechnol ; 35(7): 640-652, 2017 07.
Article in English | MEDLINE | ID: mdl-28501458

ABSTRACT

in vivo bioluminescence imaging (BLi) is an optical molecular imaging technique used to visualize molecular and cellular processes in health and diseases and to follow the fate of cells with high sensitivity using luciferase-based gene reporters. The high sensitivity of this technique arises from efficient photon production, followed by the reaction between luciferase enzymes and luciferin substrates. Novel discoveries and developments of luciferase reporters, substrates, and gene-editing techniques, and emerging fields of applications, promise a new era of deeper and more sensitive molecular imaging.


Subject(s)
Luminescent Measurements , Molecular Imaging , Firefly Luciferin/chemistry , Gene Editing/instrumentation , Gene Editing/methods , Genes, Reporter , Luciferases/chemistry , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/instrumentation , Luminescent Measurements/methods , Molecular Imaging/instrumentation , Molecular Imaging/methods
11.
Cell Discov ; 2: 16036, 2016.
Article in English | MEDLINE | ID: mdl-27924221

ABSTRACT

Generation of induced cardiomyocytes (iCMs) directly from fibroblasts offers a great opportunity for cardiac disease modeling and cardiac regeneration. A major challenge of iCM generation is the low conversion rate. To address this issue, we attempted to identify small molecules that could potentiate the reprogramming ability towards cardiac fate by removing inhibitory roadblocks. Using mouse embryonic fibroblasts as the starting cell source, we first screened 47 cardiac development related epigenetic and transcription factors, and identified an unexpected role of H3K4 methyltransferase Mll1 and related factor Men1 in inhibiting iCM reprogramming. We then applied small molecules (MM408 and MI503) of Mll1 pathway inhibitors and observed an improved efficiency in converting embryonic fibroblasts and cardiac fibroblasts into functional cardiomyocyte-like cells. We further observed that these inhibitors directly suppressed the expression of Mll1 target gene Ebf1 involved in adipocyte differentiation. Consequently, Mll1 inhibition significantly decreased the formation of adipocytes during iCM induction. Therefore, Mll1 inhibitors likely increased iCM efficiency by suppressing alternative lineage gene expression. Our studies show that targeting Mll1 dependent H3K4 methyltransferase activity provides specificity in the process of cardiac reprogramming. These findings shed new light on the molecular mechanisms underlying cardiac conversion of fibroblasts and provide novel targets and small molecules to improve iCM reprogramming for clinical applications.

12.
Cell Discov ; 2: 16008, 2016.
Article in English | MEDLINE | ID: mdl-27462455

ABSTRACT

Mixed lineage leukemia protein-1 (MLL1) has a critical role in human MLL1 rearranged leukemia (MLLr) and is a validated therapeutic target. However, its role in regulating global gene expression in MLLr cells, as well as its interplay with MLL1 fusion proteins remains unclear. Here we show that despite shared DNA-binding and cofactor interacting domains at the N terminus, MLL1 and MLL-AF9 are recruited to distinct chromatin regions and have divergent functions in regulating the leukemic transcription program. We demonstrate that MLL1, probably through C-terminal interaction with WDR5, is recruited to regulatory enhancers that are enriched for binding sites of E-twenty-six (ETS) family transcription factors, whereas MLL-AF9 binds to chromatin regions that have no H3K4me1 enrichment. Transcriptome-wide changes induced by different small molecule inhibitors also highlight the distinct functions of MLL1 and MLL-AF9. Taken together, our studies provide novel insights on how MLL1 and MLL fusion proteins contribute to leukemic gene expression, which have implications for developing effective therapies in the future.

13.
Cell Stem Cell ; 18(4): 481-94, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26996599

ABSTRACT

The interconversion between naive and primed pluripotent states is accompanied by drastic epigenetic rearrangements. However, it is unclear whether intrinsic epigenetic events can drive reprogramming to naive pluripotency or if distinct chromatin states are instead simply a reflection of discrete pluripotent states. Here, we show that blocking histone H3K4 methyltransferase MLL1 activity with the small-molecule inhibitor MM-401 reprograms mouse epiblast stem cells (EpiSCs) to naive pluripotency. This reversion is highly efficient and synchronized, with more than 50% of treated EpiSCs exhibiting features of naive embryonic stem cells (ESCs) within 3 days. Reverted ESCs reactivate the silenced X chromosome and contribute to embryos following blastocyst injection, generating germline-competent chimeras. Importantly, blocking MLL1 leads to global redistribution of H3K4me1 at enhancers and represses lineage determinant factors and EpiSC markers, which indirectly regulate ESC transcription circuitry. These findings show that discrete perturbation of H3K4 methylation is sufficient to drive reprogramming to naive pluripotency.


Subject(s)
Cellular Reprogramming/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Mouse Embryonic Stem Cells/drug effects , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Oligopeptides/pharmacology , Pluripotent Stem Cells/drug effects , Small Molecule Libraries/pharmacology , Animals , Cell Line , Germ Layers/drug effects , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/metabolism , Pluripotent Stem Cells/metabolism
14.
Mol Cell ; 53(2): 247-61, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24389101

ABSTRACT

Here we report a comprehensive characterization of our recently developed inhibitor MM-401 that targets the MLL1 H3K4 methyltransferase activity. MM-401 is able to specifically inhibit MLL1 activity by blocking MLL1-WDR5 interaction and thus the complex assembly. This targeting strategy does not affect other mixed-lineage leukemia (MLL) family histone methyltransferases (HMTs), revealing a unique regulatory feature for the MLL1 complex. Using MM-401 and its enantiomer control MM-NC-401, we show that inhibiting MLL1 methyltransferase activity specifically blocks proliferation of MLL cells by inducing cell-cycle arrest, apoptosis, and myeloid differentiation without general toxicity to normal bone marrow cells or non-MLL cells. More importantly, transcriptome analyses show that MM-401 induces changes in gene expression similar to those of MLL1 deletion, supporting a predominant role of MLL1 activity in regulating MLL1-dependent leukemia transcription program. We envision broad applications for MM-401 in basic and translational research.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Leukemia, Biphenotypic, Acute/enzymology , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mice , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/genetics , Oligopeptides/chemistry , Oligopeptides/physiology , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcriptome/drug effects
15.
J Med Chem ; 56(3): 1113-23, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23244744

ABSTRACT

Menin is an essential oncogenic cofactor for mixed lineage leukemia 1 (MLL1)-mediated leukemogenesis through its direct interaction with MLL1. Targeting the menin-MLL1 protein-protein interaction represents a promising strategy to block MLL1-mediated leukemogenesis. Employing a structure-based approach and starting from a linear MLL1 octapeptide, we have designed a class of potent macrocyclic peptidomimetic inhibitors of the menin-MLL1 interaction. The most potent macrocyclic peptidomimetic (MCP-1), 34, binds to menin with a K(i) value of 4.7 nM and is >600 times more potent than the corresponding acyclic peptide. Compound 34 is also less peptide-like and has a lower molecular weight than the initial MLL1 peptide. Therefore, compound 34 serves as a promising lead structure for the design of potent and cell-permeable inhibitors of the menin-MLL1 interaction.


Subject(s)
Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Amino Acid Sequence , Crystallography, X-Ray , Histone-Lysine N-Methyltransferase , Models, Molecular , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Binding , Protein Conformation , Proto-Oncogene Proteins/metabolism
16.
J Am Chem Soc ; 135(2): 669-82, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23210835

ABSTRACT

Mixed lineage leukemia 1 (MLL1) is a histone H3 lysine 4 (H3K4) methyltransferase, and targeting the MLL1 enzymatic activity has been proposed as a novel therapeutic strategy for the treatment of acute leukemia harboring MLL1 fusion proteins. The MLL1/WDR5 protein-protein interaction is essential for MLL1 enzymatic activity. In the present study, we designed a large number of peptidomimetics to target the MLL1/WDR5 interaction based upon -CO-ARA-NH-, the minimum binding motif derived from MLL1. Our study led to the design of high-affinity peptidomimetics, which bind to WDR5 with K(i) < 1 nM and function as potent antagonists of MLL1 activity in a fully reconstituted in vitro H3K4 methyltransferase assay. Determination of co-crystal structures of two potent peptidomimetics in complex with WDR5 establishes their structural basis for high-affinity binding to WDR5. Evaluation of one such peptidomimetic, MM-102, in bone marrow cells transduced with MLL1-AF9 fusion construct shows that the compound effectively decreases the expression of HoxA9 and Meis-1, two critical MLL1 target genes in MLL1 fusion protein mediated leukemogenesis. MM-102 also specifically inhibits cell growth and induces apoptosis in leukemia cells harboring MLL1 fusion proteins. Our study provides the first proof-of-concept for the design of small-molecule inhibitors of the WDR5/MLL1 protein-protein interaction as a novel therapeutic approach for acute leukemia harboring MLL1 fusion proteins.


Subject(s)
Drug Delivery Systems , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Peptidomimetics , Small Molecule Libraries , Binding, Competitive , Histone-Lysine N-Methyltransferase/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Models, Molecular , Myeloid-Lymphoid Leukemia Protein/drug effects , Myeloid-Lymphoid Leukemia Protein/metabolism , Peptidomimetics/chemistry , Protein Binding/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
17.
Chem Biol Drug Des ; 80(2): 237-44, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22497759

ABSTRACT

A new class of 1H-benzimidazolecarboxamidines was synthesized and evaluated for in vitro antibacterial and antifungal activities, including drug-resistant bacterial strains. The most potent compound (32) has the same ratio of anti-MRSA activity as Vancomycin (minimal inhibitory concentrations value 0.78 µg/mL). The mechanism of action for 1H-benzimidazolecarboxamidine appears to be different from existing antibacterial agents. These compounds have potential for development as a new class of potent anti-MRSA agent.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemical synthesis , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Benzimidazoles/chemical synthesis , Candida/drug effects , Candidiasis/drug therapy , Humans , Methicillin Resistance/drug effects , Staphylococcal Infections/drug therapy , Vancomycin/pharmacology
18.
J Med Chem ; 53(14): 5179-85, 2010 Jul 22.
Article in English | MEDLINE | ID: mdl-20575550

ABSTRACT

MLL1 is a histone 3 lysine 4 (H3K4) methyltransferase and a promising new cancer therapeutic target. The catalytic activity of MLL1 is regulated by the formation of a core complex consisting of MLL1, WDR5, RbBP5, and Ash2L. The interaction between WDR5 and MLL1 plays an essential role in regulation of the H3K4 methyltransferase activity of MLL1 and targeting this interaction using small molecules may represent an attractive therapeutic strategy. In this study, we have defined the essential elements in MLL1 required for its high-affinity binding to WDR5. Our data showed that the minimal elements crucial for high-affinity binding of MLL1 to WDR5 are -CO-ARA-NH- motif and two intramolecular hydrogen bonds that stabilize the conformation of this motif. Two 3-mer peptides, Ac-ARA-NH(2) and Ac-ART-NH(2), were designed based upon MLL1 and H3 sequences and achieved K(i) values of 120 and 20 nM to WDR5, respectively. Our study provides a concrete basis for the design of potent peptidomimetics and nonpeptidic compounds to inhibit MLL1 activity by targeting the MLL1 and WDR5 interaction.


Subject(s)
Histone-Lysine N-Methyltransferase/chemistry , Myeloid-Lymphoid Leukemia Protein/chemistry , Oligopeptides/chemical synthesis , Amino Acid Motifs , Binding, Competitive , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Hydrogen Bonding , Models, Molecular , Molecular Dynamics Simulation , Molecular Mimicry , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Oligopeptides/chemistry , Protein Binding , Protein Conformation
19.
Arch Pharm (Weinheim) ; 337(10): 556-62, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15476288

ABSTRACT

A series of 4-(5, 6-dichloro-1H-benzimidazol-2-yl)-N-substituted benzamides were synthesized and evaluated for antibacterial and antifungal activities against Staphylococcus aureus, methicillin-resistant S. aureus (MRSA), methicillin-resistant S. epidermis (MRSE), Enterococcus faecalis, Escherichia coli and Candida albicans. Certain compounds inhibit bacterial growth with low MIC values (microg/mL). Among them, compounds 10 and 11 exhibited the greatest antibacterial activity with MIC values of 3.12 microg/mL against S. aureus, MRSA and MRSE.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Antifungal Agents/chemical synthesis , Benzamides/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Benzamides/chemistry , Benzamides/pharmacology , Candida albicans/drug effects , Enterococcus faecalis/drug effects , Escherichia coli/drug effects , Methicillin Resistance , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...