Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Hepatology ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38954820

ABSTRACT

Alpha-1 antitrypsin deficiency (A1ATD) is a life-threatening condition caused by inheritance of the SERPINA1 'Z' genetic variant (PiZ) driving AAT protein misfolding in hepatocytes. There remain no approved medicines for this disease. Here, we report the results of a small molecule screen performed in patient derived iPSC-hepatocytes that identified Leucine-rich repeat kinase-2 (LRRK2) as a potentially new therapeutic target. Of the commercially available LRRK2 inhibitors tested, we identified CZC-25146, a candidate with favorable pharmacokinetic properties, as being capable of reducing polymer load, increasing normal AAT secretion, and reducing inflammatory cytokines in both cells and PiZ mice. Mechanistically, this effect was achieved through induction of autophagy. Our findings support the use of CZC-25146 and LRRK2 inhibitors in hepatic proteinopathy research and their further investigation as novel therapeutic candidates for A1ATD.

3.
Cell Death Dis ; 12(8): 770, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34354042

ABSTRACT

Rare monogenic disorders often share molecular etiologies involved in the pathogenesis of common diseases. Congenital disorders of glycosylation (CDG) and deglycosylation (CDDG) are rare pediatric disorders with symptoms that range from mild to life threatening. A biological mechanism shared among CDG and CDDG as well as more common neurodegenerative diseases such as Alzheimer's disease and amyotrophic lateral sclerosis, is endoplasmic reticulum (ER) stress. We developed isogenic human cellular models of two types of CDG and the only known CDDG to discover drugs that can alleviate ER stress. Systematic phenotyping confirmed ER stress and identified elevated autophagy among other phenotypes in each model. We screened 1049 compounds and scored their ability to correct aberrant morphology in each model using an agnostic cell-painting assay based on >300 cellular features. This primary screen identified multiple compounds able to correct morphological phenotypes. Independent validation shows they also correct cellular phenotypes and alleviate each of the ER stress markers identified in each model. Many of the active compounds are associated with microtubule dynamics, which points to new therapeutic opportunities for both rare and more common disorders presenting with ER stress, such as Alzheimer's disease and amyotrophic lateral sclerosis.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Models, Biological , Protective Agents/pharmacology , Activating Transcription Factor 6/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Shape/drug effects , Congenital Disorders of Glycosylation/pathology , Drug Evaluation, Preclinical , Endoplasmic Reticulum Stress/drug effects , Humans , Phenotype , Reproducibility of Results , X-Box Binding Protein 1/metabolism
4.
Circ Res ; 125(7): 699-706, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31426700

ABSTRACT

RATIONALE: Gq signaling in cardiac myocytes is classically considered toxic. Targeting Gq directly to test this is problematic, because cardiac myocytes have many Gq-coupled receptors. OBJECTIVE: Test whether Gq coupling is required for the cardioprotective effects of an alpha-1A-AR (adrenergic receptor) agonist. METHODS AND RESULTS: In recombinant cells, a mouse alpha-1A-AR with a 6-residue substitution in the third intracellular loop does not couple to Gq signaling. Here we studied a knockin mouse with this alpha-1A-AR mutation. Heart alpha-1A receptor levels and antagonist affinity in the knockin were identical to wild-type. In wild-type cardiac myocytes, the selective alpha-1A agonist A61603-stimulated phosphoinositide-phospholipase C and myocyte contraction. In myocytes with the alpha-1A knockin, both A61603 effects were absent, indicating that Gq coupling was absent. Surprisingly, A61603 activation of cardioprotective ERK (extracellular signal-regulated kinase) was markedly impaired in the KI mutant myocytes, and A61603 did not protect mutant myocytes from doxorubicin toxicity in vitro. Similarly, mice with the α1A KI mutation had increased mortality after transverse aortic constriction, and A61603 did not rescue cardiac function in mice with the Gq coupling-defective alpha-1A receptor. CONCLUSIONS: Gq coupling is required for cardioprotection by an alpha-1A-AR agonist. Gq signaling can be adaptive.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/pharmacology , Cardiotonic Agents/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Imidazoles/pharmacology , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Tetrahydronaphthalenes/pharmacology , Amino Acid Substitution , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Phosphoinositide Phospholipase C/metabolism , Protein Domains , Receptors, Adrenergic, alpha-1/chemistry , Receptors, Adrenergic, alpha-1/genetics , Signal Transduction
5.
PLoS One ; 14(6): e0217776, 2019.
Article in English | MEDLINE | ID: mdl-31158268

ABSTRACT

Friedreich's Ataxia (FA) is an inherited neurodegenerative disorder resulting from decreased expression of the mitochondrial protein frataxin, for which there is no approved therapy. High throughput screening of clinically used drugs identified Dimethyl fumarate (DMF) as protective in FA patient cells. Here we demonstrate that DMF significantly increases frataxin gene (FXN) expression in FA cell model, FA mouse model and in DMF treated humans. DMF also rescues mitochondrial biogenesis deficiency in FA-patient derived cell model. We further examined the mechanism of DMF's frataxin induction in FA patient cells. It has been shown that transcription-inhibitory R-loops form at GAA expansion mutations, thus decreasing FXN expression. In FA patient cells, we demonstrate that DMF significantly increases transcription initiation. As a potential consequence, we observe significant reduction in both R-loop formation and transcriptional pausing thereby significantly increasing FXN expression. Lastly, DMF dosed Multiple Sclerosis (MS) patients showed significant increase in FXN expression by ~85%. Since inherited deficiency in FXN is the primary cause of FA, and DMF is demonstrated to increase FXN expression in humans, DMF could be considered for Friedreich's therapy.


Subject(s)
Dimethyl Fumarate/therapeutic use , Friedreich Ataxia/drug therapy , Iron-Binding Proteins/metabolism , Animals , Dimethyl Fumarate/pharmacology , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Friedreich Ataxia/blood , Humans , Iron-Binding Proteins/blood , Iron-Binding Proteins/genetics , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Models, Biological , Mutation/genetics , Organelle Biogenesis , Transcription Initiation, Genetic , Frataxin
6.
Eur J Pharmacol ; 853: 299-307, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30965058

ABSTRACT

Transient receptor potential melastatin type 2 (TRPM2) is a cation channel activated by free intracellular ADP-ribose and reactive oxygen species. TRPM2 signaling has been linked to the pathophysiology of CNS disorders such as neuropathic pain, bipolar disorder and Alzheimer's disease. In this manuscript, we describe the discovery of JNJ-28583113, a potent brain penetrant TRPM2 antagonist. Ca2+ flux assays in cells overexpressing TRPM2 and electrophysiological recordings were used to test the pharmacology of JNJ-28583113. JNJ-28583113 was assayed in vitro on GSK-3 phosphorylation levels, cell death, cytokine release in microglia and unbiased morphological phenotypic analysis. Finally, we dosed animals to evaluate its pharmacokinetic properties. Our results showed that JNJ-28583113 is a potent (126 ±â€¯0.5 nM) TRPM2 antagonist. Blocking TRPM2 caused phosphorylation of GSK3α and ß subunits. JNJ-28583113 also protected cells from oxidative stress induced cell death as well as morphological changes induced by non-cytotoxic concentrations of H2O2. In addition, inhibiting TRPM2 blunted cytokine release in response to pro-inflammatory stimuli in microglia. Lastly, we showed that JNJ-28583113 was brain penetrant but not suitable for systemic dosing as it was rapidly metabolized in vivo. While the in-vitro pharmacology of JNJ-28583113 is the best in class, its in-vivo properties would need optimization to assist in further probing key roles of TRPM2 in CNS pathophysiology.


Subject(s)
Drug Discovery , Pyrazoles/pharmacology , TRPM Cation Channels/antagonists & inhibitors , Animals , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Rats
7.
Environ Health Perspect ; 125(8): 087015, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28885978

ABSTRACT

BACKGROUND: Quaternary ammonium salts (QUATS), such as cetylpyridinium chloride (CPC) and benzalkonium chloride (BAK), are frequently used in antiseptic formulations, including toothpastes, mouthwashes, lozenges, throat and nasal sprays, and as biocides. Although in a recent ruling, the U.S. Food and Drug Administration (FDA) banned CPC from certain products and requested more data on BAK's efficacy and safety profile, QUATS, in general, and CPC and BAK, in particular, continue to be used in personal health care, food, and pharmaceutical and cleaning industries. OBJECTIVES: We aimed to assess CPC's effects on mitochondrial toxicity and endocrine disruption in vitro. METHOD: Mitochondrial O2 consumption and adenosine triphosphate (ATP) synthesis rates of osteosarcoma cybrid cells were measured before and after CPC and BAK treatment. Antiestrogenic effects of the compounds were measured by a luciferase-based assay using recombinant human breast carcinoma cells (VM7Luc4E2, ERalpha-positive). RESULTS: CPC inhibited both mitochondrial O2 consumption [half maximal inhibitory concentration (IC50): 3.8µM] and ATP synthesis (IC50: 0.9µM), and additional findings supported inhibition of mitochondrial complex 1 as the underlying mechanism for these effects. In addition, CPC showed concentration-dependent antiestrogenic activity half maximal effective concentration [(EC50): 4.5µM)]. BAK, another antimicrobial QUATS that is structurally similar to CPC, and the pesticide rotenone, a known complex 1 inhibitor, also showed mitochondrial inhibitory and antiestrogenic effects. In all three cases, there was overlap of the antiestrogenic activity with the mitochondrial inhibitory activity. CONCLUSIONS: Mitochondrial inhibition in vitro occurred at a CPC concentration that may be relevant to human exposures. The antiestrogenic activity of CPC, BAK, rotenone, and triclosan may be related to their mitochondrial inhibitory activity. Our findings support the need for additional research on the mitochondrial inhibitory and antiestrogenic effects of QUATS, including CPC and BAK. https://doi.org/10.1289/EHP1404.


Subject(s)
Anti-Infective Agents, Local/toxicity , Cetylpyridinium/toxicity , Cell Line, Tumor , Humans , Mitochondria/drug effects
8.
Hum Mol Genet ; 26(15): 2864-2873, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28460056

ABSTRACT

The induction of mitochondrial biogenesis could potentially alleviate mitochondrial and muscle disease. We show here that dimethyl fumarate (DMF) dose-dependently induces mitochondrial biogenesis and function dosed to cells in vitro, and also dosed in vivo to mice and humans. The induction of mitochondrial gene expression is more dependent on DMF's target Nrf2 than hydroxycarboxylic acid receptor 2 (HCAR2). Thus, DMF induces mitochondrial biogenesis primarily through its action on Nrf2, and is the first drug demonstrated to increase mitochondrial biogenesis with in vivo human dosing. This is the first demonstration that mitochondrial biogenesis is deficient in Multiple Sclerosis patients, which could have implications for MS pathophysiology and therapy. The observation that DMF stimulates mitochondrial biogenesis, gene expression and function suggests that it could be considered for mitochondrial disease therapy and/or therapy in muscle disease in which mitochondrial function is important.


Subject(s)
Dimethyl Fumarate , NF-E2-Related Factor 2 , Animals , Humans , Mice , Cell Culture Techniques , Dimethyl Fumarate/chemistry , Dimethyl Fumarate/metabolism , Fibroblasts , Mitochondria/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neuroprotective Agents/pharmacology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Organelle Biogenesis
9.
Mitochondrion ; 31: 79-83, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27717841

ABSTRACT

Mitochondrial toxicity is emerging as a major mechanism underlying serious human health consequences. This work performs a high-throughput screen (HTS) of 176 environmental chemicals for mitochondrial toxicity utilizing a previously reported biosensor platform. This established HTS confirmed known mitochondrial toxins and identified novel mitotochondrial uncouplers such as 2, 2'-Methylenebis(4-chlorophenol) and pentachlorophenol. It also identified a mitochondrial 'structure activity relationship' (SAR) in the sense that multiple environmental chlorophenols are mitochondrial inhibitors and uncouplers. This study demonstrates proof-of-concept that a mitochondrial HTS assay detects known and novel environmental mitotoxicants, and could be used to quickly evaluate human health risks from mitotoxicants in the environment.


Subject(s)
Biosensing Techniques/methods , Environmental Pollutants/toxicity , High-Throughput Screening Assays/methods , Mitochondria/drug effects , Mitochondria/metabolism , Toxicology/methods , Cell Line , Energy Metabolism/drug effects , Epithelial Cells/drug effects , Epithelial Cells/physiology , Humans , Neurons/drug effects , Neurons/physiology , Uncoupling Agents/toxicity
10.
PLoS One ; 10(1): e0115701, 2015.
Article in English | MEDLINE | ID: mdl-25606852

ABSTRACT

The α1A-AR is thought to couple predominantly to the Gαq/PLC pathway and lead to phosphoinositide hydrolysis and calcium mobilization, although certain agonists acting at this receptor have been reported to trigger activation of arachidonic acid formation and MAPK pathways. For several G protein-coupled receptors (GPCRs) agonists can manifest a bias for activation of particular effector signaling output, i.e., not all agonists of a given GPCR generate responses through utilization of the same signaling cascade(s). Previous work with Gαq coupling-defective variants of α1A-AR, as well as a combination of Ca2+ channel blockers, uncovered cross-talk between α1A-AR and ß2-AR that leads to potentiation of a Gαq-independent signaling cascade in response to α1A-AR activation. We hypothesized that molecules exist that act as biased agonists to selectively activate this pathway. In this report, isoproterenol (Iso), typically viewed as ß-AR-selective agonist, was examined with respect to activation of α1A-AR. α1A-AR selective antagonists were used to specifically block Iso evoked signaling in different cellular backgrounds and confirm its action at α1A-AR. Iso induced signaling at α1A-AR was further interrogated by probing steps along the Gαq /PLC, Gαs and MAPK/ERK pathways. In HEK-293/EBNA cells transiently transduced with α1A-AR, and CHO_α1A-AR stable cells, Iso evoked low potency ERK activity as well as Ca2+ mobilization that could be blocked by α1A-AR selective antagonists. The kinetics of Iso induced Ca2+ transients differed from typical Gαq- mediated Ca2+ mobilization, lacking both the fast IP3R mediated response and the sustained phase of Ca2+ re-entry. Moreover, no inositol phosphate (IP) accumulation could be detected in either cell line after stimulation with Iso, but activation was accompanied by receptor internalization. Data are presented that indicate that Iso represents a novel type of α1A-AR partial agonist with signaling bias toward MAPK/ERK signaling cascade that is likely independent of coupling to Gαq.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/pharmacology , Calcium Signaling/drug effects , Isoproterenol/pharmacology , MAP Kinase Signaling System/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Animals , CHO Cells , Calcium Signaling/genetics , Cricetulus , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , HEK293 Cells , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , MAP Kinase Signaling System/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Adrenergic, alpha-1/genetics
11.
Aging Cell ; 13(6): 1049-58, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25257068

ABSTRACT

Adipose tissue is an important metabolic organ that integrates a wide array of homeostatic processes and is crucial for whole-body insulin sensitivity and energy metabolism. Brown adipose tissue (BAT) is a key thermogenic tissue with a well-established role in energy expenditure. BAT dissipates energy and protects against both hypothermia and obesity. Thus, BAT stimulation therapy is a rational strategy for the looming pandemic of obesity, whose consequences and comorbidities have a huge impact on the aged. Shc-deficient mice (ShcKO) were previously shown to be lean, insulin sensitive, and resistant to high-fat diet and obesity. We investigated the contribution of BAT to this phenotype. Insulin-dependent BAT glucose uptake was higher in ShcKO mice. Primary ShcKO BAT cells exhibited increased mitochondrial respiration; increased expression of several mitochondrial and lipid-oxidative enzymes was observed in ShcKO BAT. Levels of brown fat-specific markers of differentiation, UCP1, PRDM16, ELOVL3, and Cox8b, were higher in ShcKO BAT. In vitro, Shc knockdown in BAT cell line increased insulin sensitivity and metabolic activity. In vivo, pharmacological stimulation of ShcKO BAT resulted in higher energy expenditure. Conversely, pharmacological inhibition of BAT abolished the improved metabolic parameters, that is the increased insulin sensitivity and glucose tolerance of ShcKO mice. Similarly, in vitro Shc knockdown in BAT cell lines increased their expression of UCP1 and metabolic activity. These data suggest increased BAT activity significantly contributes to the improved metabolic phenotype of ShcKO mice.


Subject(s)
Adipose Tissue, Brown/metabolism , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/metabolism , Animals , Energy Metabolism , Mice , Mice, Knockout , Thermogenesis/physiology
12.
Hum Mol Genet ; 23(25): 6848-62, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25113747

ABSTRACT

Inherited deficiency in the mitochondrial protein frataxin (FXN) causes the rare disease Friedreich's ataxia (FA), for which there is no successful treatment. We identified a redox deficiency in FA cells and used this to model the disease. We screened a 1600-compound library to identify existing drugs, which could be of therapeutic benefit. We identified the topical anesthetic dyclonine as protective. Dyclonine increased FXN transcript and FXN protein dose-dependently in FA cells and brains of animal models. Dyclonine also rescued FXN-dependent enzyme deficiencies in the iron-sulfur enzymes, aconitase and succinate dehydrogenase. Dyclonine induces the Nrf2 [nuclear factor (erythroid-derived 2)-like 2] transcription factor, which we show binds an upstream response element in the FXN locus. Additionally, dyclonine also inhibited the activity of histone methyltransferase G9a, known to methylate histone H3K9 to silence FA chromatin. Chronic dosing in a FA mouse model prevented a performance decline in balance beam studies. A human clinical proof-of-concept study was completed in eight FA patients dosed twice daily using a 1% dyclonine rinse for 1 week. Six of the eight patients showed an increase in buccal cell FXN levels, and fold induction was significantly correlated with disease severity. Dyclonine represents a novel therapeutic strategy that can potentially be repurposed for the treatment of FA.


Subject(s)
Anesthetics, Local/pharmacology , Friedreich Ataxia/drug therapy , Iron-Binding Proteins/agonists , Mouth Mucosa/drug effects , NF-E2-Related Factor 2/agonists , Neuroprotective Agents/pharmacology , Propiophenones/pharmacology , Aconitate Hydratase/genetics , Aconitate Hydratase/metabolism , Animals , Cell Line , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/pathology , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Gene Expression Regulation , High-Throughput Screening Assays , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Postural Balance/drug effects , Signal Transduction , Small Molecule Libraries/pharmacology , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Frataxin
13.
Mitochondrion ; 17: 116-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25034306

ABSTRACT

Repurposing of FDA-approved drugs with effects on mitochondrial function might shorten the critical path to mitochondrial disease drug development. We improved a biosensor-based assay of mitochondrial O2 consumption, and identified mitofunctional defects in cell models of LHON and FXTAS. Using this platform, we screened a 1600-compound library of clinically used drugs. The assay identified drugs known to affect mitochondrial function, such as metformin and decoquinate. We also identified several drugs not previously known to affect mitochondrial respiration including acarbose, metaraminol, gallamine triethiodide, and acamprosate. These previously unknown 'mitoactives' represent novel links to targets for mitochondrial regulation and potentially therapy, for mitochondrial disease.


Subject(s)
Biosensing Techniques , Cell Respiration/drug effects , Drug Discovery/methods , Mitochondria/drug effects , Oxygen/analysis , High-Throughput Screening Assays/methods , Humans , Mitochondria/metabolism
14.
J Pharmacol Exp Ther ; 350(2): 455-68, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24917542

ABSTRACT

Both preclinical evidence and clinical evidence suggest that α7 nicotinic acetylcholine receptor activation (α7nAChR) improves cognitive function, the decline of which is associated with conditions such as Alzheimer's disease and schizophrenia. Moreover, allosteric modulation of α7nAChR is an emerging therapeutic strategy in an attempt to avoid the rapid desensitization properties associated with the α7nAChR after orthosteric activation. We used a calcium assay to screen for positive allosteric modulators (PAMs) of α7nAChR and report on the pharmacologic characterization of the novel compound RO5126946 (5-chloro-N-[(1S,3R)-2,2-dimethyl-3-(4-sulfamoyl-phenyl)-cyclopropyl]-2-methoxy-benzamide), which allosterically modulates α7nAChR activity. RO5126946 increased acetylcholine-evoked peak current and delayed current decay but did not affect the recovery of α7nAChRs from desensitization. In addition, RO5126946's effects were absent when nicotine-evoked currents were completely blocked by coapplication of the α7nAChR-selective antagonist methyl-lycaconitine. RO5126946 enhanced α7nAChR synaptic transmission and positively modulated GABAergic responses. The absence of RO5126946 effects at human α4ß2nAChR and 5-hydroxytryptamine 3 receptors, among others, indicated selectivity for α7nAChRs. In vivo, RO5126946 is orally bioavailable and brain-penetrant and improves associative learning in a scopolamine-induced deficit model of fear conditioning in rats. In addition, procognitive effects of RO5126946 were investigated in the presence of nicotine to address potential pharmacologic interactions on behavior. RO5126946 potentiated nicotine's effects on fear memory when both compounds were administered at subthreshold doses and did not interfere with procognitive effects observed when both compounds were administered at effective doses. Overall, RO5126946 is a novel α7nAChR PAM with cognitive-enhancing properties.


Subject(s)
Benzamides/pharmacology , Sulfonamides/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/drug effects , Allosteric Regulation , Animals , Cells, Cultured , Cognition/drug effects , Hippocampus/drug effects , Humans , Learning/drug effects , Male , Memory/drug effects , Nicotine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/physiology , Receptors, Glutamate/physiology
15.
Bioorg Med Chem ; 21(8): 2346-2354, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23473946

ABSTRACT

The effect of the alkyl side chain length of coenzyme Q10 on mitochondrial respiratory chain function has been investigated by the use of synthetic ubiquinone derivatives. Three analogues (3, 4 and 6) were identified that exhibited significantly improved effects on mitochondrial oxygen consumption and mitochondrial membrane potential, and also conferred significant cytoprotection on cultured mammalian cells in which glutathione had been depleted by treatment with diethyl maleate. The analogues also exhibited lesser inhibition of the electron transport chain than idebenone. The results obtained provide guidance for the design of CoQ10 analogues with improved activity compared to that of idebenone (1), the latter of which is undergoing evaluation in the clinic as a therapeutic agent.


Subject(s)
Electron Transport/drug effects , Mitochondria/drug effects , Ubiquinone/analogs & derivatives , Animals , Cattle , Cell Line , Cell Line, Tumor , Cytoprotection , Electron Transport/physiology , Humans , Leukemia/metabolism , Leukemia/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Structure-Activity Relationship , Ubiquinone/chemistry , Ubiquinone/metabolism , Ubiquinone/pharmacology
16.
Anal Biochem ; 434(1): 105-11, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23219563

ABSTRACT

The EPXH2 gene encodes soluble epoxide hydrolase (sEH), which has two distinct enzyme activities: epoxide hydrolase (Cterm-EH) and phosphatase (Nterm-phos). The Cterm-EH is involved in the metabolism of arachidonic acid epoxides that play important roles in blood pressure, cell growth, inflammation, and pain. While recent findings suggested complementary biological roles for Nterm-phos, research is limited by the lack of potent bioavailable inhibitors of this phosphatase activity. Also, a potent bioavailable inhibitor of this activity could be important in the development of therapy for cardiovascular diseases. We report herein the development of an HTS enzyme-based assay for Nterm-phos (Z'>0.9) using AttoPhos as the substrate. This assay was used to screen a wide variety of chemical entities, including a library of known drugs that have reached through clinical evaluation (Pharmakon 1600), as well as a library of pesticides and environmental toxins. We discovered that ebselen inhibits sEH phosphatase activity. Ebselen binds to the N-terminal domain of sEH (K(I)=550 nM) and chemically reacts with the enzyme to quickly and irreversibly inhibit Nterm-phos, and subsequently Cterm-EH, and thus represents a new class of sEH inhibitor.


Subject(s)
Epoxide Hydrolases/analysis , High-Throughput Screening Assays , Small Molecule Libraries/chemistry , Azoles/metabolism , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Humans , Isoindoles , Kinetics , Organoselenium Compounds/metabolism , Pesticides/metabolism , Protein Binding , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Substrate Specificity , Toxins, Biological/metabolism
17.
Mol Pharmacol ; 75(3): 713-28, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19109357

ABSTRACT

Agonist occupied alpha(1)-adrenoceptors (alpha(1)-ARs) engage several signaling pathways, including phosphatidylinositol hydrolysis, calcium mobilization, arachidonic acid release, mitogen-activated protein (MAP) kinase activation, and cAMP accumulation. The natural agonist norepinephrine (NE) activates with variable affinity and intrinsic efficacy all adrenoceptors, and in cells that coexpress alpha(1)- and beta-AR subtypes, such as cardiomyocytes, this leads to coactivation of multiple downstream pathways. This may result in pathway cross-talk with significant consequences to heart physiology and pathologic state. To dissect signaling components involved specifically in alpha(1A)- and beta(2)-AR signal interplay, we have developed a recombinant model system that mimics the levels of receptor expression observed in native cells. We followed intracellular Ca(2+) mobilization to monitor in real time the activation of both G(q) and G(s) pathways. We found that coactivation of alpha(1A)- and beta(2)-AR by the nonselective agonist NE or via a combination of the highly selective alpha(1A)-AR agonist A61603 and the beta-selective agonist isoproterenol led to increases in Ca(2+) influx from the extracellular compartment relative to stimulation with A61603 alone, with no effect on the associated transient release of Ca(2+) from intracellular stores. This effect became more evident upon examination of an alpha(1A)-AR variant exhibiting a partial defect in coupling to G(q), and we attribute it to potentiation of a non G(q)-pathway, uncovered by application of a combination of xestospongin C, an endoplasmic reticulum inositol 1,4,5-triphosphate receptor blocker, and 2-aminoethoxydiphenyl borate, a nonselective storeoperated Ca(2+) entry channel blocker. We also found that stimulation with A61603 of a second alpha(1A)-AR variant entirely unable to signal induced no Ca(2+) unless beta(2)-AR was concomitantly activated. These results may be accounted for by the presence of alpha(1A)/beta(2)-AR heterodimers or alternatively by specific adrenoceptor signal cross-talk resulting in distinct pharmacological behavior. Finally, our findings provide a new conceptual framework to rationalize outcomes from clinical studies targeting alpha- and beta-adrenoceptors.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Intracellular Membranes/physiology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta-2/physiology , Signal Transduction/physiology , Amino Acid Sequence , Cell Line , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Intracellular Membranes/pathology , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...