Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
iScience ; 27(2): 108968, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38327788

ABSTRACT

Excessive or aberrant NLRP3 inflammasome activation has been implicated in the progression and initiation of many inflammatory conditions; however, currently no NLRP3 inflammasome inhibitors have been approved for therapeutic use in the clinic. Here we have identified that the natural product brazilin effectively inhibits both priming and activation of the NLRP3 inflammasome in cultured murine macrophages, a human iPSC microglial cell line and in a mouse model of acute peritoneal inflammation. Through computational modeling, we predict that brazilin can adopt a favorable binding pose within a site of the NLRP3 protein which is essential for its conformational activation. Our results not only encourage further evaluation of brazilin as a therapeutic agent for NLRP3-related inflammatory diseases, but also introduce this small-molecule as a promising scaffold structure for the development of derivative NLRP3 inhibitor compounds.

2.
J Med Chem ; 63(5): 2308-2324, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31430136

ABSTRACT

The lysyl oxidase (LOX) family of extracellular proteins plays a vital role in catalyzing the formation of cross-links in fibrillar elastin and collagens leading to extracellular matrix (ECM) stabilization. These enzymes have also been implicated in tumor progression and metastatic disease and have thus become an attractive therapeutic target for many types of invasive cancers. Following our recently published work on the discovery of aminomethylenethiophenes (AMTs) as potent, orally bioavailable LOX/LOXL2 inhibitors, we report herein the discovery of a series of dual LOX/LOXL2 inhibitors, as well as a subseries of LOXL2-selective inhibitors, bearing an aminomethylenethiazole (AMTz) scaffold. Incorporation of a thiazole core leads to improved potency toward LOXL2 inhibition via an irreversible binding mode of inhibition. SAR studies have enabled the discovery of a predictive 3DQSAR model. Lead AMTz inhibitors exhibit improved pharmacokinetic properties and excellent antitumor efficacy, with significantly reduced tumor growth in a spontaneous breast cancer genetically engineered mouse model.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Thiazoles/pharmacology , Amination , Amino Acid Oxidoreductases/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Female , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/pathology , Protein-Lysine 6-Oxidase/metabolism , Rats , Sulfinic Acids/chemistry , Sulfinic Acids/pharmacokinetics , Sulfinic Acids/pharmacology , Sulfinic Acids/therapeutic use , Thiazoles/chemistry , Thiazoles/pharmacokinetics , Thiazoles/therapeutic use
3.
J Med Chem ; 62(12): 5863-5884, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31070916

ABSTRACT

Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase that cross-links collagens and elastin in the extracellular matrix and is a critical mediator of tumor growth and metastatic spread. LOX is a target for cancer therapy, and thus the search for therapeutic agents against LOX has been widely sought. We report herein the medicinal chemistry discovery of a series of LOX inhibitors bearing an aminomethylenethiophene (AMT) scaffold. High-throughput screening provided the initial hits. Structure-activity relationship (SAR) studies led to the discovery of AMT inhibitors with sub-micromolar half-maximal inhibitory concentrations (IC50) in a LOX enzyme activity assay. Further SAR optimization yielded the orally bioavailable LOX inhibitor CCT365623 with good anti-LOX potency, selectivity, pharmacokinetic properties, as well as anti-metastatic efficacy.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biological Availability , Cell Line, Tumor , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Humans , Mice , Neoplasm Metastasis/drug therapy , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/pharmacokinetics , Thiophenes/pharmacology , Thiophenes/therapeutic use
4.
Development ; 145(5)2018 03 12.
Article in English | MEDLINE | ID: mdl-29530939

ABSTRACT

During ontogeny, hematopoietic stem and progenitor cells arise from hemogenic endothelium through an endothelial-to-hematopoietic transition that is strictly dependent on the transcription factor RUNX1. Although it is well established that RUNX1 is essential for the onset of hematopoiesis, little is known about the role of RUNX1 dosage specifically in hemogenic endothelium and during the endothelial-to-hematopoietic transition. Here, we used the mouse embryonic stem cell differentiation system to determine if and how RUNX1 dosage affects hemogenic endothelium differentiation. The use of inducible Runx1 expression combined with alterations in the expression of the RUNX1 co-factor CBFß allowed us to evaluate a wide range of RUNX1 levels. We demonstrate that low RUNX1 levels are sufficient and necessary to initiate an effective endothelial-to-hematopoietic transition. Subsequently, RUNX1 is also required to complete the endothelial-to-hematopoietic transition and to generate functional hematopoietic precursors. In contrast, elevated levels of RUNX1 are able to drive an accelerated endothelial-to-hematopoietic transition, but the resulting cells are unable to generate mature hematopoietic cells. Together, our results suggest that RUNX1 dosage plays a pivotal role in hemogenic endothelium maturation and the establishment of the hematopoietic system.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Endothelium, Vascular/physiology , Gene Dosage/physiology , Hemangioblasts/physiology , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/physiology , Gene Expression Regulation , Mice , Mice, Knockout
5.
PLoS One ; 12(7): e0181902, 2017.
Article in English | MEDLINE | ID: mdl-28732076

ABSTRACT

Long non-coding RNAs are being increasingly recognised as important molecules involved in regulating a diverse array of biological functions. For example, many long non-coding RNAs have been associated with tumourigenesis and in this context their molecular functions often involves impacting on chromatin and transcriptional control processes. One important cellular control system that is often deregulated in cancer cells is the ERK MAP kinase pathway. Here we have investigated whether ERK pathway signaling in response to EGF stimulation, leads to changes in the production of long non-coding RNAs. We identify several different classes of EGF pathway-regulated lncRNAs. We focus on one of the inducible lincRNAs, EGF inducible long intergenic non-coding RNA 1 (EINCR1). EINCR1 is predominantly nuclear and shows delayed activation kinetics compared to other immediate-early EGF-inducible genes. In humans it is expressed in a tissue-specific manner and is mainly confined to the heart but it exhibits little evolutionary conservation. Importantly, in several cancers EINCR1 shows elevated expression levels which correlate with poor survival in lung adenocarcinoma patients. In the context of lung adenocarcinomas, EINCR1 expression is anti-correlated with the expression of several protein coding EGF-regulated genes. A potential functional connection is demonstrated as EINCR1 overexpression is shown to reduce the expression of EGF-regulated protein coding genes including FOS and FOSB.


Subject(s)
Adenocarcinoma/genetics , Epidermal Growth Factor/genetics , Gene Expression Regulation/genetics , Lung Neoplasms/genetics , RNA, Long Noncoding/genetics , A549 Cells , Adenocarcinoma of Lung , Cell Line , Cell Line, Tumor , Chromatin/genetics , HEK293 Cells , Humans , MAP Kinase Signaling System/genetics
6.
Dev Cell ; 36(5): 572-87, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26923725

ABSTRACT

Metazoan development involves the successive activation and silencing of specific gene expression programs and is driven by tissue-specific transcription factors programming the chromatin landscape. To understand how this process executes an entire developmental pathway, we generated global gene expression, chromatin accessibility, histone modification, and transcription factor binding data from purified embryonic stem cell-derived cells representing six sequential stages of hematopoietic specification and differentiation. Our data reveal the nature of regulatory elements driving differential gene expression and inform how transcription factor binding impacts on promoter activity. We present a dynamic core regulatory network model for hematopoietic specification and demonstrate its utility for the design of reprogramming experiments. Functional studies motivated by our genome-wide data uncovered a stage-specific role for TEAD/YAP factors in mammalian hematopoietic specification. Our study presents a powerful resource for studying hematopoiesis and demonstrates how such data advance our understanding of mammalian development.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Animals , Cell Lineage/physiology , Homeodomain Proteins/metabolism , Mice , Protein Binding/genetics , Transcription Factors/metabolism
7.
J Med Chem ; 58(11): 4790-801, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25977981

ABSTRACT

The RAS/RAF/MEK/ERK signaling pathway has been targeted with a number of small molecule inhibitors in oncology clinical development across multiple disease indications. Importantly, cell lines with acquired resistance to B-RAF and MEK inhibitors have been shown to maintain sensitivity to ERK1/2 inhibition by small molecule inhibitors. There are a number of selective, noncovalent ERK1/2 inhibitors reported along with the promiscuous hypothemycin (and related analogues) that act via a covalent mechanism of action. This article reports the identification of multiple series of highly selective covalent ERK1/2 inhibitors informed by structure-based drug design (SBDD). As a starting point for these covalent inhibitors, reported ERK1/2 inhibitors and a chemical series identified via high-throughput screening were exploited. These approaches resulted in the identification of selective covalent tool compounds for potential in vitro and in vivo studies to assess the risks and or benefits of targeting this pathway through such a mechanism of action.


Subject(s)
Drug Design , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 3/chemistry , Protein Kinase Inhibitors/pharmacology , Amino Acid Sequence , Cells, Cultured , Crystallography, X-Ray , Humans , Immunoblotting , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
8.
J Biomol Screen ; 20(6): 748-59, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25851036

ABSTRACT

Here, we describe an approach to identify novel selective estrogen receptor downregulator (SERD) compounds with improved properties such as oral bioavailability and the potential of increased efficacy compared to currently marketed drug treatments. Previously, methodologies such as Western blotting and transient cell reporter assays have been used to identify and characterize SERD compounds, but such approaches can be limited due to low throughput and sensitivity, respectively. We have used an endogenous cell-imaging strategy that has both the throughput and sensitivity to support a large-scale hit-to-lead program to identify novel compounds. A screening cascade with a suite of assays has been developed to characterize compounds that modulate estrogen receptor α (ERα)-mediated signaling or downregulate ERα levels in cells. Initially, from a focused high-throughput screening, novel ERα binders were identified that could be modified chemically into ERα downregulators. Following this, cellular assays helped determine the mechanism of action of compounds to distinguish between on-target and off-target compounds and differentiate SERDs, selective estrogen receptor modulator (SERM) compounds, and agonist ERα ligands. Data are shown to exemplify the characterization of ERα-mediated signaling inhibitors using a selection of literature compounds and illustrate how this cascade has been used to drive the chemical design of novel SERD compounds.


Subject(s)
Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Down-Regulation , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , High-Throughput Screening Assays , Humans , Protein Binding , Receptors, Progesterone/metabolism , Small Molecule Libraries
SELECTION OF CITATIONS
SEARCH DETAIL
...