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.
J Med Chem ; 65(10): 7380-7398, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35549469

ABSTRACT

Inhibitors of Kelch-like ECH-associated protein 1 (Keap1) increase the activity of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) by stalling its ubiquitination and degradation. This enhances the expression of genes encoding proteins involved in drug detoxification, redox homeostasis, and mitochondrial function. Nrf2 activation offers a potential therapeutic approach for conditions including Alzheimer's and Parkinson's diseases, vascular inflammation, and chronic obstructive airway disease. Non-electrophilic Keap1-Nrf2 protein-protein interaction (PPI) inhibitors may have improved toxicity profiles and different pharmacological properties to cysteine-reactive electrophilic inhibitors. Here, we describe and characterize a series of phenyl bis-sulfonamide PPI inhibitors that bind to Keap1 at submicromolar concentrations. Structural studies reveal that the compounds bind to Keap1 in a distinct "peptidomimetic" conformation that resembles the Keap1-Nrf2 ETGE peptide complex. This is different to other small molecule Keap1-Nrf2 PPI inhibitors, including bicyclic aryl bis-sulfonamides, offering a starting point for new design approaches to Keap1 inhibitors.


Subject(s)
NF-E2-Related Factor 2 , Sulfonamides , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Protein Binding , Sulfonamides/pharmacology
2.
Chembiochem ; 19(17): 1810-1816, 2018 09 04.
Article in English | MEDLINE | ID: mdl-29927029

ABSTRACT

Noncovalent inhibitors of the Keap1-Nrf2 protein-protein interaction (PPI) have therapeutic potential in a range of disease states including neurodegenerative diseases (Parkinson's and Alzheimer's diseases), chronic obstructive pulmonary disease and various inflammatory conditions. By stalling Keap1-mediated ubiquitination of Nrf2, such compounds can enhance Nrf2 transcriptional activity and activate the expression of a range of genes with antioxidant response elements in their promoter regions. Keap1 inhibitors based on peptide and small-molecule templates have been identified. In this paper we develop the structure-activity relationships of the peptide series and identify a group of ligands incorporating unnatural amino acids that demonstrate improved binding affinity in fluorescence polarisation, differential scanning fluorimetry and isothermal titration calorimetry assays. These modified peptides have the potential for further development into peptidomimetic chemical probes to explore the role of Nrf2 in disease and as potential lead structures for drug development.


Subject(s)
Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , NF-E2-Related Factor 2/antagonists & inhibitors , Oligopeptides/chemistry , Protein Binding/drug effects , Amino Acids/chemistry , Binding Sites/drug effects , Crystallography, X-Ray , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Molecular Structure , NF-E2-Related Factor 2/metabolism , Oligopeptides/chemical synthesis , Protein Conformation , Protein Domains , Structure-Activity Relationship , Thermodynamics
3.
Cell Death Dis ; 8(6): e2896, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28640253

ABSTRACT

The 18 kDa translocator protein TSPO localizes on the outer mitochondrial membrane (OMM). Systematically overexpressed at sites of neuroinflammation it is adopted as a biomarker of brain conditions. TSPO inhibits the autophagic removal of mitochondria by limiting PARK2-mediated mitochondrial ubiquitination via a peri-organelle accumulation of reactive oxygen species (ROS). Here we describe that TSPO deregulates mitochondrial Ca2+ signaling leading to a parallel increase in the cytosolic Ca2+ pools that activate the Ca2+-dependent NADPH oxidase (NOX) thereby increasing ROS. The inhibition of mitochondrial Ca2+ uptake by TSPO is a consequence of the phosphorylation of the voltage-dependent anion channel (VDAC1) by the protein kinase A (PKA), which is recruited to the mitochondria, in complex with the Acyl-CoA binding domain containing 3 (ACBD3). Notably, the neurotransmitter glutamate, which contributes neuronal toxicity in age-dependent conditions, triggers this TSPO-dependent mechanism of cell signaling leading to cellular demise. TSPO is therefore proposed as a novel OMM-based pathway to control intracellular Ca2+ dynamics and redox transients in neuronal cytotoxicity.


Subject(s)
Calcium/metabolism , Homeostasis , Mitochondria/metabolism , Receptors, GABA/metabolism , Signal Transduction , Stress, Physiological , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Glutamic Acid/pharmacology , Homeostasis/drug effects , Humans , Mice , Mitochondria/drug effects , Mitochondrial Membranes/metabolism , Models, Biological , NADPH Oxidases/metabolism , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Stress, Physiological/drug effects , Voltage-Dependent Anion Channels/metabolism
4.
PLoS Genet ; 13(3): e1006593, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28253260

ABSTRACT

Nrf2, a transcriptional activator of cell protection genes, is an attractive therapeutic target for the prevention of neurodegenerative diseases, including Alzheimer's disease (AD). Current Nrf2 activators, however, may exert toxicity and pathway over-activation can induce detrimental effects. An understanding of the mechanisms mediating Nrf2 inhibition in neurodegenerative conditions may therefore direct the design of drugs targeted for the prevention of these diseases with minimal side-effects. Our study provides the first in vivo evidence that specific inhibition of Keap1, a negative regulator of Nrf2, can prevent neuronal toxicity in response to the AD-initiating Aß42 peptide, in correlation with Nrf2 activation. Comparatively, lithium, an inhibitor of the Nrf2 suppressor GSK-3, prevented Aß42 toxicity by mechanisms independent of Nrf2. A new direct inhibitor of the Keap1-Nrf2 binding domain also prevented synaptotoxicity mediated by naturally-derived Aß oligomers in mouse cortical neurons. Overall, our findings highlight Keap1 specifically as an efficient target for the re-activation of Nrf2 in AD, and support the further investigation of direct Keap1 inhibitors for the prevention of neurodegeneration in vivo.


Subject(s)
Alzheimer Disease/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Profiling/methods , Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Animals, Genetically Modified , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Lithium Chloride/pharmacology , Longevity/drug effects , Longevity/genetics , Mice , Microscopy, Confocal , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neurons/metabolism , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Binding/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Thiadiazoles/pharmacology , Triazoles/pharmacology
5.
Biochem J ; 473(2): 107-21, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26733718

ABSTRACT

The 18-kDa translocator protein (TSPO) localizes in the outer mitochondrial membrane (OMM) of cells and is readily up-regulated under various pathological conditions such as cancer, inflammation, mechanical lesions and neurological diseases. Able to bind with high affinity synthetic and endogenous ligands, its core biochemical function resides in the translocation of cholesterol into the mitochondria influencing the subsequent steps of (neuro-)steroid synthesis and systemic endocrine regulation. Over the years, however, TSPO has also been linked to core cellular processes such as apoptosis and autophagy. It interacts and forms complexes with other mitochondrial proteins such as the voltage-dependent anion channel (VDAC) via which signalling and regulatory transduction of these core cellular events may be influenced. Despite nearly 40 years of study, the precise functional role of TSPO beyond cholesterol trafficking remains elusive even though the recent breakthroughs on its high-resolution crystal structure and contribution to quality-control signalling of mitochondria. All this along with a captivating pharmacological profile provides novel opportunities to investigate and understand the significance of this highly conserved protein as well as contribute the development of specific therapeutics as presented and discussed in the present review.


Subject(s)
Mitochondria/metabolism , Receptors, GABA/genetics , Receptors, GABA/metabolism , Amino Acid Sequence , Animals , Biochemical Phenomena , Cholesterol/metabolism , Drug Delivery Systems/trends , Humans , Mitochondrial Membranes/metabolism , Molecular Sequence Data , Steroids/administration & dosage , Steroids/metabolism
6.
Biochem Soc Trans ; 43(4): 543-52, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26551691

ABSTRACT

The mitochondrial 18-kDa translocator protein (TSPO) was originally discovered as a peripheral binding site of benzodiazepines to be later described as a core element of cholesterol trafficking between cytosol and mitochondria from which the current nomenclature originated. The high affinity it exhibits with chemicals (i.e. PK11195) has generated interest in the development of mitochondrial based TSPO-binding drugs for in vitro and in vivo analysis. Increased TSPO expression is observed in numerous pathologies such as cancer and inflammatory conditions of the central nervous system (CNS) that have been successfully exploited via protocols of positron emission tomography (PET) imaging. We endeavoured to dissect the molecular role of TSPO in mitochondrial cell biology and discovered a functional link with quality control mechanisms operated by selective autophagy. This review focuses on the current understanding of this pathway and focuses on the interplay with reactive oxygen species (ROS) and the voltage-dependent anion channel (VDAC), to which TSPO binds, in the regulation of cell mitophagy and hence homoeostasis of the mitochondrial network as a whole.


Subject(s)
Mitophagy , Oxidation-Reduction , Receptors, GABA/metabolism , Humans , Reactive Oxygen Species/metabolism , Voltage-Dependent Anion Channels/metabolism
7.
Autophagy ; 10(12): 2279-96, 2014.
Article in English | MEDLINE | ID: mdl-25470454

ABSTRACT

The 18-kDa TSPO (translocator protein) localizes on the outer mitochondrial membrane (OMM) and participates in cholesterol transport. Here, we report that TSPO inhibits mitochondrial autophagy downstream of the PINK1-PARK2 pathway, preventing essential ubiquitination of proteins. TSPO abolishes mitochondrial relocation of SQSTM1/p62 (sequestosome 1), and consequently that of the autophagic marker LC3 (microtubule-associated protein 1 light chain 3), thus leading to an accumulation of dysfunctional mitochondria, altering the appearance of the network. Independent of cholesterol regulation, the modulation of mitophagy by TSPO is instead dependent on VDAC1 (voltage-dependent anion channel 1), to which TSPO binds, reducing mitochondrial coupling and promoting an overproduction of reactive oxygen species (ROS) that counteracts PARK2-mediated ubiquitination of proteins. These data identify TSPO as a novel element in the regulation of mitochondrial quality control by autophagy, and demonstrate the importance for cell homeostasis of its expression ratio with VDAC1.


Subject(s)
Autophagy/physiology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Receptors, GABA/metabolism , Ubiquitination/physiology , Voltage-Dependent Anion Channel 1/metabolism , Animals , Biological Transport/physiology , Mice , Mitochondrial Membranes/metabolism , Ubiquitin-Protein Ligases/metabolism
8.
Br J Pharmacol ; 171(18): 4193-206, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24641180

ABSTRACT

BACKGROUND AND PURPOSE: Ischaemia compromises mitochondrial respiration. Consequently, the mitochondrial F1 Fo-ATPsynthase reverses and acts as a proton-pumping ATPase, so maintaining the mitochondrial membrane potential (ΔΨm ), while accelerating ATP depletion and cell death. Here we have looked for a molecule that can selectively inhibit this activity without affecting ATP synthesis, preserve ATP and delay ischaemic cell death. EXPERIMENTAL APPROACH: We developed a chemoinformatic screen based on the structure of BMS199264, which is reported to selectively inhibit F1 Fo-ATPase activity and which is cardioprotective. Results suggested the molecule BTB06584 (hereafter referred to as BTB). Fluorescence microscopy was used to study its effects on ΔΨm and on the rate of ATP consumption following inhibition of respiration in several cell types. The effect of BTB on oxygen (O2 ) consumption was explored and protective potential determined using ischaemia/reperfusion assays. We also investigated a potential mechanism of action through its interaction with inhibitor protein of F1 subunit (IF1 ), the endogenous inhibitor of the F1 Fo-ATPase. KEY RESULTS: BTB inhibited F1 Fo-ATPase activity with no effect on ΔΨm or O2 consumption. ATP consumption was decreased following inhibition of respiration, and ischaemic cell death was reduced. BTB efficiency was increased by IF1 overexpression and reduced by silencing the protein. In addition, BTB rescued defective haemoglobin synthesis in zebrafish pinotage (pnt) mutants in which expression of the Atpif1a gene is lost. CONCLUSIONS AND IMPLICATIONS: BTB may represent a valuable tool to selectively inhibit mitochondrial F1 Fo-ATPase activity without compromising ATP synthesis and to limit ischaemia-induced injury caused by reversal of the mitochondrial F1 Fo-ATPsynthase.


Subject(s)
Chlorobenzoates/pharmacology , Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Proteins/metabolism , Proton-Translocating ATPases/antagonists & inhibitors , Sulfones/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Death/drug effects , Cell Line , Embryo, Nonmammalian , HeLa Cells , Hemoglobins/metabolism , Hemolysis/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neurons/drug effects , Oxygen Consumption , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Zebrafish , ATPase Inhibitory Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...