Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 50
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
ACS Pharmacol Transl Sci ; 4(3): 1136-1148, 2021 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-34151204

RÉSUMÉ

Niemann-Pick disease type C1 (NPC1) is a rare genetic cholesterol storage disorder caused by mutations in the NPC1 gene. Mutations in this transmembrane late endosome protein lead to loss of normal cholesterol efflux from late endosomes and lysosomes. It has been shown that broad spectrum histone deacetylase inhibitors (HDACi's) such as Vorinostat correct the cholesterol accumulation phenotype in the majority of NPC1 mutants tested in cultured cells. In order to determine the optimal specificity for HDACi correction of the mutant NPC1s, we screened 76 HDACi's of varying specificity. We tested the ability of these HDACi's to correct the excess accumulation of cholesterol in patient fibroblast cells that homozygously express NPC1 I1061T , the most common mutation. We determined that inhibition of HDACs 1, 2, and 3 is important for correcting the defect, and combined inhibition of all three is needed to achieve the greatest effect, suggesting a need for multiple effects of the HDACi treatments. Identifying the specific HDACs involved in the process of regulating cholesterol trafficking in NPC1 will help to focus the search for more specific druggable targets.

2.
Sci Rep ; 11(1): 9018, 2021 04 27.
Article de Anglais | MEDLINE | ID: mdl-33907245

RÉSUMÉ

Histone/protein deacetylases (HDAC) 1 and 2 are typically viewed as structurally and functionally similar enzymes present within various co-regulatory complexes. We tested differential effects of these isoforms in renal ischemia reperfusion injury (IRI) using inducible knockout mice and found no significant change in ischemic tolerance with HDAC1 deletion, but mitigation of ischemic injury with HDAC2 deletion. Restriction of HDAC2 deletion to the kidney via transplantation or PAX8-controlled proximal renal tubule-specific Cre resulted in renal IRI protection. Pharmacologic inhibition of HDAC2 increased histone acetylation in the kidney but did not extend renal protection. Protein analysis demonstrated increased HDAC1-associated CoREST protein in HDAC2-/- versus WT cells, suggesting that in the absence of HDAC2, increased CoREST complex occupancy of HDAC1 can stabilize this complex. In vivo administration of a CoREST inhibitor exacerbated renal injury in WT mice and eliminated the benefit of HDAC2 deletion. Gene expression analysis of endothelin showed decreased endothelin levels in HDAC2 deletion. These data demonstrate that contrasting effects of HDAC1 and 2 on CoREST complex stability within renal tubules can affect outcomes of renal IRI and implicate endothelin as a potential downstream mediator.


Sujet(s)
Protéines corépressives/métabolisme , Histone Deacetylase 2/métabolisme , Tubules contournés proximaux/métabolisme , Lésion d'ischémie-reperfusion/prévention et contrôle , Animaux , Protéines corépressives/antagonistes et inhibiteurs , Endothélines/métabolisme , Antienzymes/pharmacologie , Femelle , Délétion de gène , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Histone Deacetylase 2/antagonistes et inhibiteurs , Histone Deacetylase 2/génétique , Isoenzymes/antagonistes et inhibiteurs , Isoenzymes/métabolisme , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Mâle , Souris , Souris knockout
3.
Int J Mol Sci ; 21(21)2020 Oct 29.
Article de Anglais | MEDLINE | ID: mdl-33137873

RÉSUMÉ

Selective inhibition of histone deacetylase 3 (HDAC3) prevents glucolipotoxicity-induced ß-cell dysfunction and apoptosis by alleviation of proapoptotic endoplasmic reticulum (ER) stress-signaling, but the precise molecular mechanisms of alleviation are unexplored. By unbiased microarray analysis of the ß-cell gene expression profile of insulin-producing cells exposed to glucolipotoxicity in the presence or absence of a selective HDAC3 inhibitor, we identified Enhancer of zeste homolog 2 (EZH2) as the sole target candidate. ß-Cells were protected against glucolipotoxicity-induced ER stress and apoptosis by EZH2 attenuation. Small molecule inhibitors of EZH2 histone methyltransferase activity rescued human islets from glucolipotoxicity-induced apoptosis. Moreover, EZH2 knockdown cells were protected against glucolipotoxicity-induced downregulation of the protective non-canonical Nuclear factor of kappa light polypeptide gene enhancer in B-cells (NFκB) pathway. We conclude that EZH2 deficiency protects from glucolipotoxicity-induced ER stress, apoptosis and downregulation of the non-canonical NFκB pathway, but not from insulin secretory dysfunction. The mechanism likely involves transcriptional regulation via EZH2 functioning as a methyltransferase and/or as a methylation-dependent transcription factor.


Sujet(s)
Apoptose , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Glucose/effets indésirables , Sécrétion d'insuline/effets des médicaments et des substances chimiques , Cellules à insuline/anatomopathologie , Lipides/effets indésirables , Cellules cultivées , Protéine-2 homologue de l'activateur de Zeste/génétique , Humains , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Transduction du signal , Édulcorants/effets indésirables
4.
Sci Transl Med ; 12(544)2020 05 20.
Article de Anglais | MEDLINE | ID: mdl-32434848

RÉSUMÉ

Fragile X syndrome is caused by FMR1 gene silencing and loss of the encoded fragile X mental retardation protein (FMRP), which binds to mRNA and regulates translation. Studies in the Fmr1-/y mouse model of fragile X syndrome indicate that aberrant cerebral protein synthesis downstream of metabotropic glutamate receptor 5 (mGluR5) signaling contributes to disease pathogenesis, but clinical trials using mGluR5 inhibitors were not successful. Animal studies suggested that treatment with lithium might be an alternative approach. Targets of lithium include paralogs of glycogen synthase kinase 3 (GSK3), and nonselective small-molecule inhibitors of these enzymes improved disease phenotypes in a fragile X syndrome mouse model. However, the potential therapeutic use of GSK3 inhibitors has been hampered by toxicity arising from inhibition of both α and ß paralogs. Recently, we developed GSK3 inhibitors with sufficient paralog selectivity to avoid a known toxic consequence of dual inhibition, that is, increased ß-catenin stabilization. We show here that inhibition of GSK3α, but not GSK3ß, corrected aberrant protein synthesis, audiogenic seizures, and sensory cortex hyperexcitability in Fmr1-/y mice. Although inhibiting either paralog prevented induction of NMDA receptor-dependent long-term depression (LTD) in the hippocampus, only inhibition of GSK3α impaired mGluR5-dependent and protein synthesis-dependent LTD. Inhibition of GSK3α additionally corrected deficits in learning and memory in Fmr1-/y mice; unlike mGluR5 inhibitors, there was no evidence of tachyphylaxis or enhanced psychotomimetic-induced hyperlocomotion. GSK3α selective inhibitors may have potential as a therapeutic approach for treating fragile X syndrome.


Sujet(s)
Syndrome du chromosome X fragile , Animaux , Modèles animaux de maladie humaine , Protéine du syndrome X fragile/génétique , Protéine du syndrome X fragile/métabolisme , Syndrome du chromosome X fragile/traitement médicamenteux , Glycogen Synthase Kinase 3 , Hippocampe/métabolisme , Souris , Souris de lignée C57BL , Souris knockout
5.
J Cell Biochem ; 121(1): 244-258, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31222845

RÉSUMÉ

The regulation of epigenetic factors is an emerging therapeutic target of immune function in a variety of osteolytic pathologies. Histone deacetylases (HDAC) modify core histone proteins and transcriptional processes, in addition to nonhistone protein activity. The activated immune response in rheumatoid arthritis, periodontitis, and prosthetic implant particle release stimulates the catabolic activity of osteoclasts. In this study, we investigated the effects of novel therapeutic agents targeting HDAC isozymes (HDAC 1, 2, and 5), previously shown to be upregulated in inflammatory bone disorders, in cytokine-stimulated human monocytes and osteoclasts in vitro. Inhibiting HDAC 1 and 2 significantly reduced gene expression of IL-1ß, TNF, MCP-1, and MIP-1α in TNF-stimulated monocytes, while suppressing secretions of IL-1ß, IL-10, INF-γ, and MCP-1 (P < .05). Osteoclast formation and bone resorption were also significantly diminished with HDAC 1 and 2 inhibition, through reduced NFATc1 expression and osteoclast specific target genes, TRAF6, CTR, TRAP, and Cathepsin K (P < .05). Similar trends were observed when inhibiting HDAC 1 and to a lesser extent, HDAC 2, in isolation. However, their combined inhibition had the greatest anti-inflammatory and antiosteoclastic effects. Targeting HDAC 5 had minimal effects on these processes investigated in this study, whereas a broad acting HDACi, 1179.4b, had widespread suppressive outcomes. This study demonstrates that targeting HDACs is a potent and effective way of regulating the inflammatory and catabolic processes in human monocytes and osteoclasts. It also demonstrates the importance of targeting individual HDACs with an overall aim to improve efficiency and reduce any potential off target effects.


Sujet(s)
Résorption osseuse , Cytokines/métabolisme , Antienzymes/pharmacologie , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 2/antagonistes et inhibiteurs , Ostéoclastes/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Chimiokines/métabolisme , Histone Deacetylase 1/métabolisme , Histone Deacetylase 2/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Histone deacetylases/métabolisme , Humains , Inflammation , Agranulocytes/métabolisme , Monocytes/métabolisme , Ostéoclastes/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(10): 1545-1561, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31051283

RÉSUMÉ

Niemann-Pick type C1 (NPC1) disease is a fatal neurovisceral disease for which there are no FDA approved treatments, though cyclodextrin (HPßCD) slows disease progression in preclinical models and in an early phase clinical trial. Our goal was to evaluate the mechanism of action of a previously described combination-therapy, Triple Combination Formulation (TCF) - comprised of the histone deacetylase inhibitor (HDACi) vorinostat/HPßCD/PEG - shown to prolong survival in Npc1 mice. In these studies, TCF's benefit was attributed to enhanced vorinostat pharmacokinetics (PK). Here, we show that TCF reduced lipid storage, extended lifespan, and preserved neurological function in Npc1 mice. Unexpectedly, substitution of an inactive analog for vorinostat in TCF revealed similar efficacy. We demonstrate that the efficacy of TCF was attributable to enhanced HPßCD PK and independent of NPC1 protein expression. We conclude that although HDACi effectively reduce cholesterol storage in NPC1-deficient cells, HDACi are ineffective in vivo in Npc1 mice.


Sujet(s)
2-Hydroxypropyl-beta-cyclodextrin/usage thérapeutique , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Maladie de Niemann-Pick de type C/traitement médicamenteux , Polyéthylène glycols/usage thérapeutique , Vorinostat/usage thérapeutique , Animaux , Cellules cultivées , Association médicamenteuse , Protéines et peptides de signalisation intracellulaire/métabolisme , Mâle , Souris , Souris de lignée BALB C , Protéine NPC1 , Maladie de Niemann-Pick de type C/métabolisme
7.
Neuropsychopharmacology ; 44(6): 1152-1162, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-30647450

RÉSUMÉ

Chronic stress promotes depression in some individuals, but has no effect in others. Susceptible individuals exhibit social avoidance and anxious behavior and ultimately develop depression, whereas resilient individuals live normally. Exercise counteracts the effects of stress. Our objective was to examine whether lactate, a metabolite produced during exercise and known to reproduce specific brain exercise-related changes, promotes resilience to stress and acts as an antidepressant. To determine whether lactate promotes resilience to stress, male C57BL/6 mice experienced daily defeat by a CD-1 aggressor, for 10 days. On the 11th day, mice were subjected to behavioral tests. Mice received lactate before each defeat session. When compared with control mice, mice exposed to stress displayed increased susceptibility, social avoidance and anxiety. Lactate promoted resilience to stress and rescued social avoidance and anxiety by restoring hippocampal class I histone deacetylase (HDAC) levels and activity, specifically HDAC2/3. To determine whether lactate is an antidepressant, mice only received lactate from days 12-25 and a second set of behavioral tests was conducted on day 26. In this paradigm, we examined whether lactate functions by regulating HDACs using co-treatment with CI-994, a brain-permeable class I HDAC inhibitor. When administered after the establishment of depression, lactate behaved as antidepressant. In this paradigm, lactate regulated HDAC5 and not HDAC2/3 levels. On the contrary, HDAC2/3 inhibition was antidepressant-like. This indicates that lactate mimics exercise's effects and rescues susceptibility to stress by modulating HDAC2/3 activity and suggests that HDAC2/3 play opposite roles before and after establishment of susceptibility to stress.


Sujet(s)
Antidépresseurs/pharmacologie , Anxiété/prévention et contrôle , Apprentissage par évitement , Dépression/métabolisme , Hippocampe/métabolisme , Histone deacetylases/métabolisme , Acide lactique/pharmacologie , Résilience psychologique , Comportement social , Stress psychologique/prévention et contrôle , Animaux , Antidépresseurs/administration et posologie , Apprentissage par évitement/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Benzamides , Dépression/traitement médicamenteux , Modèles animaux de maladie humaine , Prédisposition aux maladies , Hippocampe/effets des médicaments et des substances chimiques , Histone Deacetylase 2/effets des médicaments et des substances chimiques , Histone Deacetylase 2/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Histone deacetylases/effets des médicaments et des substances chimiques , Acide lactique/administration et posologie , Mâle , Souris , Souris de lignée C57BL , Phénylènediamines/pharmacologie , Résilience psychologique/effets des médicaments et des substances chimiques
8.
Sci Transl Med ; 10(431)2018 03 07.
Article de Anglais | MEDLINE | ID: mdl-29515000

RÉSUMÉ

Glycogen synthase kinase 3 (GSK3), a key regulatory kinase in the wingless-type MMTV integration site family (WNT) pathway, is a therapeutic target of interest in many diseases. Although dual GSK3α/ß inhibitors have entered clinical trials, none has successfully translated to clinical application. Mechanism-based toxicities, driven in part by the inhibition of both GSK3 paralogs and subsequent ß-catenin stabilization, are a concern in the translation of this target class because mutations and overexpression of ß-catenin are associated with many cancers. Knockdown of GSK3α or GSK3ß individually does not increase ß-catenin and offers a conceptual resolution to targeting GSK3: paralog-selective inhibition. However, inadequate chemical tools exist. The design of selective adenosine triphosphate (ATP)-competitive inhibitors poses a drug discovery challenge due to the high homology (95% identity and 100% similarity) in this binding domain. Taking advantage of an Asp133→Glu196 "switch" in their kinase hinge, we present a rational design strategy toward the discovery of paralog-selective GSK3 inhibitors. These GSK3α- and GSK3ß-selective inhibitors provide insights into GSK3 targeting in acute myeloid leukemia (AML), where GSK3α was identified as a therapeutic target using genetic approaches. The GSK3α-selective compound BRD0705 inhibits kinase function and does not stabilize ß-catenin, mitigating potential neoplastic concerns. BRD0705 induces myeloid differentiation and impairs colony formation in AML cells, with no apparent effect on normal hematopoietic cells. Moreover, BRD0705 impairs leukemia initiation and prolongs survival in AML mouse models. These studies demonstrate feasibility of paralog-selective GSK3α inhibition, offering a promising therapeutic approach in AML.


Sujet(s)
Antienzymes/usage thérapeutique , Glycogen Synthase Kinase 3/antagonistes et inhibiteurs , Leucémie aigüe myéloïde/traitement médicamenteux , Dipeptides/composition chimique , Dipeptides/métabolisme , Glycogen Synthase Kinase 3/composition chimique , Glycogen Synthase Kinase 3/métabolisme , Humains , Mutagenèse dirigée , Isoformes de protéines/antagonistes et inhibiteurs , Isoformes de protéines/composition chimique , Isoformes de protéines/métabolisme , Cellules U937 , bêta-Caténine/génétique , bêta-Caténine/métabolisme
9.
ACS Chem Biol ; 13(4): 1038-1047, 2018 04 20.
Article de Anglais | MEDLINE | ID: mdl-29485852

RÉSUMÉ

Schizophrenia is a severe neuropsychiatric disease that lacks completely effective and safe therapies. As a polygenic disorder, genetic studies have only started to shed light on its complex etiology. To date, the positive symptoms of schizophrenia are well-managed by antipsychotic drugs, which primarily target the dopamine D2 receptor (D2R). However, these antipsychotics are often accompanied by severe side effects, including motoric symptoms. At D2R, antipsychotic drugs antagonize both G-protein dependent (Gαi/o) signaling and G-protein independent (ß-arrestin) signaling. However, the relevant contributions of the distinct D2R signaling pathways to antipsychotic efficacy and on-target side effects (motoric) are still incompletely understood. Recent evidence from mouse genetic and pharmacological studies point to ß-arrestin signaling as the major driver of antipsychotic efficacy and suggest that a ß-arrestin biased D2R antagonist could achieve an additional level of selectivity at D2R, increasing the therapeutic index of next generation antipsychotics. Here, we characterize BRD5814, a highly brain penetrant ß-arrestin biased D2R antagonist. BRD5814 demonstrated good target engagement via PET imaging, achieving efficacy in an amphetamine-induced hyperlocomotion mouse model with strongly reduced motoric side effects in a rotarod performance test. This proof of concept study opens the possibility for the development of a new generation of pathway selective antipsychotics at D2R with reduced side effect profiles for the treatment of schizophrenia.


Sujet(s)
Neuroleptiques/usage thérapeutique , Récepteur D2 de la dopamine/effets des médicaments et des substances chimiques , bêta-Arrestines/métabolisme , Animaux , Imagerie diagnostique/méthodes , Protéines G/antagonistes et inhibiteurs , Humains , Locomotion/effets des médicaments et des substances chimiques , Souris , Schizophrénie/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , bêta-Arrestines/antagonistes et inhibiteurs
10.
J Biol Chem ; 292(43): 17598-17608, 2017 10 27.
Article de Anglais | MEDLINE | ID: mdl-28860191

RÉSUMÉ

Preservation of insulin-secreting ß-cells is an important goal for therapies aimed at restoring normoglycemia in patients with diabetes. One approach, the inhibition of histone deacetylases (HDACs), has been reported to suppress pancreatic islet inflammation and ß-cell apoptosis in vitro In this report, we demonstrate the efficacy of HDAC inhibitors (HDACi) in vivo We show that daily administration of BRD3308, an isoform-selective HDAC3 inhibitor, for 2 weeks to female nonobese diabetic (NOD) mice, beginning at 3 weeks of age, followed by twice-weekly injections until age 25 weeks, protects the animals from diabetes. The preservation of ß-cells was because of a significant decrease in islet infiltration of mononuclear cells. Moreover, the BRD3308 treatment increased basal insulin secretion from islets cultured in vitro All metabolic tissues tested in vehicle- or BRD3308-treated groups showed virtually no sign of immune cell infiltration, except minimal infiltration in white adipose tissue in animals treated with the highest BRD3308 dose (10 mg/kg), providing additional evidence of protection from immune attack in the treated groups. Furthermore, pancreata from animals treated with 10 mg/kg BRD3308 exhibited significantly decreased numbers of apoptotic ß-cells compared with those treated with vehicle or low-dose BRD3308. Finally, animals treated with 1 or 10 mg/kg BRD3308 had enhanced ß-cell proliferation. These in vivo results point to the potential use of selective HDAC3 inhibitors as a therapeutic approach to suppress pancreatic islet infiltration and prevent ß-cell death with the long-term goal of limiting the progression of type 1 diabetes.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Diabète expérimental/prévention et contrôle , Inhibiteurs de désacétylase d'histone/pharmacologie , Histone deacetylases/métabolisme , Cellules à insuline/enzymologie , Tissu adipeux blanc/enzymologie , Tissu adipeux blanc/anatomopathologie , Animaux , Diabète expérimental/enzymologie , Diabète expérimental/anatomopathologie , Femelle , Cellules à insuline/anatomopathologie , Souris , Souris de lignée NOD
11.
Sci Signal ; 10(493)2017 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-28831019

RÉSUMÉ

Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), have emerged as key regulators of brain plasticity and represent disease-modifying targets for several brain disorders, including Alzheimer's disease and major depressive disorder. Because of poor pharmacokinetic properties of BDNF, the interest in small-molecule TrkB agonists and modulators is high. Several compounds have been reported to act as TrkB agonists, and their increasing use in various nervous system disorder models creates the perception that these are reliable probes. To examine key pharmacological parameters of these compounds in detail, we have developed and optimized a series of complementary quantitative assays that measure TrkB receptor activation, TrkB-dependent downstream signaling, and gene expression in different cellular contexts. Although BDNF and other neurotrophic factors elicited robust and dose-dependent receptor activation and downstream signaling, we were unable to reproduce these activities using the reported small-molecule TrkB agonists. Our findings indicate that experimental results obtained with these compounds must be carefully interpreted and highlight the challenge of developing reliable pharmacological activators of this key molecular target.


Sujet(s)
Facteur neurotrophique dérivé du cerveau/métabolisme , Glycoprotéines membranaires/agonistes , Neuroblastome/traitement médicamenteux , Neurones/effets des médicaments et des substances chimiques , Récepteur trkB/agonistes , Transduction du signal , Bibliothèques de petites molécules/pharmacologie , Animaux , Cellules cultivées , Embryon de mammifère/cytologie , Embryon de mammifère/effets des médicaments et des substances chimiques , Embryon de mammifère/métabolisme , Test ELISA , Flavanones/pharmacologie , Tests de criblage à haut débit , Humains , Glycoprotéines membranaires/métabolisme , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Neurones/cytologie , Neurones/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Rats , Récepteur trkB/métabolisme
12.
J Pharmacol Exp Ther ; 361(1): 140-150, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28174211

RÉSUMÉ

Inhibitors of zinc-dependent histone deacetylases (HDACs) profoundly affect cellular function by altering gene expression via changes in nucleosomal histone tail acetylation. Historically, investigators have employed pan-HDAC inhibitors, such as the hydroxamate trichostatin A (TSA), which simultaneously targets members of each of the three zinc-dependent HDAC classes (classes I, II, and IV). More recently, class- and isoform-selective HDAC inhibitors have been developed, providing invaluable chemical biology probes for dissecting the roles of distinct HDACs in the control of various physiologic and pathophysiological processes. For example, the benzamide class I HDAC-selective inhibitor, MGCD0103 [N-(2-aminophenyl)-4-[[(4-pyridin-3-ylpyrimidin-2-yl)amino]methyl] benzamide], was shown to block cardiac fibrosis, a process involving excess extracellular matrix deposition, which often results in heart dysfunction. Here, we compare the mechanisms of action of structurally distinct HDAC inhibitors in isolated primary cardiac fibroblasts, which are the major extracellular matrix-producing cells of the heart. TSA, MGCD0103, and the cyclic peptide class I HDAC inhibitor, apicidin, exhibited a common ability to enhance histone acetylation, and all potently blocked cardiac fibroblast cell cycle progression. In contrast, MGCD0103, but not TSA or apicidin, paradoxically increased expression of a subset of fibrosis-associated genes. Using the cellular thermal shift assay, we provide evidence that the divergent effects of HDAC inhibitors on cardiac fibroblast gene expression relate to differential engagement of HDAC1- and HDAC2-containing complexes. These findings illustrate the importance of employing multiple compounds when pharmacologically assessing HDAC function in a cellular context and during HDAC inhibitor drug development.


Sujet(s)
Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/enzymologie , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/pharmacologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/enzymologie , Animaux , Animaux nouveau-nés , Cellules cultivées , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/métabolisme , Souris , Souris de lignée C57BL , Rats , Rat Sprague-Dawley
13.
Cancer Discov ; 7(1): 38-53, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27733359

RÉSUMÉ

Somatic mutations in CREBBP occur frequently in B-cell lymphoma. Here, we show that loss of CREBBP facilitates the development of germinal center (GC)-derived lymphomas in mice. In both human and murine lymphomas, CREBBP loss-of-function resulted in focal depletion of enhancer H3K27 acetylation and aberrant transcriptional silencing of genes that regulate B-cell signaling and immune responses, including class II MHC. Mechanistically, CREBBP-regulated enhancers are counter-regulated by the BCL6 transcriptional repressor in a complex with SMRT and HDAC3, which we found to bind extensively to MHC class II loci. HDAC3 loss-of-function rescued repression of these enhancers and corresponding genes, including MHC class II, and more profoundly suppressed CREBBP-mutant lymphomas in vitro and in vivo Hence, CREBBP loss-of-function contributes to lymphomagenesis by enabling unopposed suppression of enhancers by BCL6/SMRT/HDAC3 complexes, suggesting HDAC3-targeted therapy as a precision approach for CREBBP-mutant lymphomas. SIGNIFICANCE: Our findings establish the tumor suppressor function of CREBBP in GC lymphomas in which CREBBP mutations disable acetylation and result in unopposed deacetylation by BCL6/SMRT/HDAC3 complexes at enhancers of B-cell signaling and immune response genes. Hence, inhibition of HDAC3 can restore the enhancer histone acetylation and may serve as a targeted therapy for CREBBP-mutant lymphomas. Cancer Discov; 7(1); 38-53. ©2016 AACR.See related commentary by Höpken, p. 14This article is highlighted in the In This Issue feature, p. 1.


Sujet(s)
Protéine CBP/génétique , Centre germinatif/métabolisme , Histone deacetylases/génétique , Lymphome B diffus à grandes cellules/génétique , Mutation , Acétylation , Animaux , Protéine CBP/métabolisme , Lignée cellulaire tumorale , Éléments activateurs (génétique) , Techniques de knock-out de gènes , Histone deacetylases/métabolisme , Histone/métabolisme , Humains , Lymphome B diffus à grandes cellules/métabolisme , Souris , Transplantation tumorale , Corépresseur-2 de récepteur nucléaire/génétique , Protéines proto-oncogènes c-bcl-6/génétique , Transcription génétique
14.
Future Med Chem ; 8(17): 2033-2046, 2016 Nov.
Article de Anglais | MEDLINE | ID: mdl-27739327

RÉSUMÉ

AIM: Caffeic acid (3,4-dihydroxycinnamic acid) phenethyl ester (CAPE), the major constituent of propolis, is able to increase the survival of the nematode Caenorhabditis elegans after infection with the fungal pathogen Candida albicans. RESULTS: CAPE increases the expression of several antimicrobial proteins involved in the immune response to C. albicans. Structural derivatives of CAPE were synthesized to identify structure-activity relationships and decrease metabolic liability, ultimately leading to a compound that has similar efficacy, but increased in vivo stability. The CED-10(Rac-1)/PAK1 pathway was essential for immunomodulation by CAPE and was a critical component involved in the immune response to fungal pathogens. CONCLUSION: Caenorhabditis elegans is an efficient heterologous host to evaluate immunomodulatory compounds and identify components of the pathway(s) involved in the mode of action of compounds.

15.
Bioorg Med Chem ; 24(18): 4008-4015, 2016 09 15.
Article de Anglais | MEDLINE | ID: mdl-27377864

RÉSUMÉ

The structure-activity and structure-kinetic relationships of a series of novel and selective ortho-aminoanilide inhibitors of histone deacetylases (HDACs) 1 and 2 are described. Different kinetic and thermodynamic selectivity profiles were obtained by varying the moiety occupying an 11Å channel leading to the Zn(2+) catalytic pocket of HDACs 1 and 2, two paralogs with a high degree of structural similarity. The design of these novel inhibitors was informed by two ligand-bound crystal structures of truncated hHDAC2. BRD4884 and BRD7232 possess kinetic selectivity for HDAC1 versus HDAC2. We demonstrate that the binding kinetics of HDAC inhibitors can be tuned for individual isoforms in order to modulate target residence time while retaining functional activity and increased histone H4K12 and H3K9 acetylation in primary mouse neuronal cell culture assays. These chromatin modifiers, with tuned binding kinetic profiles, can be used to define the relation between target engagement requirements and the pharmacodynamic response of HDACs in different disease applications.


Sujet(s)
Anilides/composition chimique , Anilides/pharmacologie , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 2/antagonistes et inhibiteurs , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/pharmacologie , Acétylation/effets des médicaments et des substances chimiques , Amination , Animaux , Cellules cultivées , Histone Deacetylase 1/métabolisme , Histone Deacetylase 2/métabolisme , Histone/métabolisme , Humains , Cinétique , Souris , Simulation de docking moléculaire
16.
Angew Chem Int Ed Engl ; 55(33): 9601-5, 2016 08 08.
Article de Anglais | MEDLINE | ID: mdl-27355874

RÉSUMÉ

Glycogen synthase kinase-3 (GSK-3) regulates multiple cellular processes in diabetes, oncology, and neurology. N-(3-(1H-1,2,4-triazol-1-yl)propyl)-5-(3-chloro-4-methoxyphenyl)oxazole-4-carboxamide (PF-04802367 or PF-367) has been identified as a highly potent inhibitor, which is among the most selective antagonists of GSK-3 to date. Its efficacy was demonstrated in modulation of tau phosphorylation in vitro and in vivo. Whereas the kinetics of PF-367 binding in brain tissues are too fast for an effective therapeutic agent, the pharmacokinetic profile of PF-367 is ideal for discovery of radiopharmaceuticals for GSK-3 in the central nervous system. A (11) C-isotopologue of PF-367 was synthesized and preliminary PET imaging studies in non-human primates confirmed that we have overcome the two major obstacles for imaging GSK-3, namely, reasonable brain permeability and displaceable binding.


Sujet(s)
Encéphale/effets des médicaments et des substances chimiques , Encéphale/imagerie diagnostique , Neuroimagerie , Oxazoles/pharmacologie , Tomographie par émission de positons , Inhibiteurs de protéines kinases/pharmacologie , Triazoles/pharmacologie , Protéines tau/antagonistes et inhibiteurs , Encéphale/métabolisme , Cristallographie aux rayons X , Relation dose-effet des médicaments , Glycogen Synthase Kinase 3/antagonistes et inhibiteurs , Glycogen Synthase Kinase 3/métabolisme , Humains , Modèles moléculaires , Structure moléculaire , Oxazoles/synthèse chimique , Oxazoles/composition chimique , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Triazoles/synthèse chimique , Triazoles/composition chimique , Protéines tau/métabolisme
17.
Elife ; 52016 06 02.
Article de Anglais | MEDLINE | ID: mdl-27253067

RÉSUMÉ

Exercise induces beneficial responses in the brain, which is accompanied by an increase in BDNF, a trophic factor associated with cognitive improvement and the alleviation of depression and anxiety. However, the exact mechanisms whereby physical exercise produces an induction in brain Bdnf gene expression are not well understood. While pharmacological doses of HDAC inhibitors exert positive effects on Bdnf gene transcription, the inhibitors represent small molecules that do not occur in vivo. Here, we report that an endogenous molecule released after exercise is capable of inducing key promoters of the Mus musculus Bdnf gene. The metabolite ß-hydroxybutyrate, which increases after prolonged exercise, induces the activities of Bdnf promoters, particularly promoter I, which is activity-dependent. We have discovered that the action of ß-hydroxybutyrate is specifically upon HDAC2 and HDAC3, which act upon selective Bdnf promoters. Moreover, the effects upon hippocampal Bdnf expression were observed after direct ventricular application of ß-hydroxybutyrate. Electrophysiological measurements indicate that ß-hydroxybutyrate causes an increase in neurotransmitter release, which is dependent upon the TrkB receptor. These results reveal an endogenous mechanism to explain how physical exercise leads to the induction of BDNF.


Sujet(s)
Acide 3-hydroxy-butyrique/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Acétylation , Animaux , Facteur neurotrophique dérivé du cerveau/génétique , Cellules cultivées , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Histone Deacetylase 2/antagonistes et inhibiteurs , Histone deacetylases/composition chimique , Histone/métabolisme , Mâle , Souris , Souris de lignée C57BL , Neurones/cytologie , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Agents neuromédiateurs/métabolisme , Conditionnement physique d'animal , Récepteur trkB/métabolisme
18.
ACS Chem Biol ; 11(7): 1952-63, 2016 07 15.
Article de Anglais | MEDLINE | ID: mdl-27128528

RÉSUMÉ

The mood stabilizer lithium, the first-line treatment for bipolar disorder, is hypothesized to exert its effects through direct inhibition of glycogen synthase kinase 3 (GSK3) and indirectly by increasing GSK3's inhibitory serine phosphorylation. GSK3 comprises two highly similar paralogs, GSK3α and GSK3ß, which are key regulatory kinases in the canonical Wnt pathway. GSK3 stands as a nodal target within this pathway and is an attractive therapeutic target for multiple indications. Despite being an active field of research for the past 20 years, many GSK3 inhibitors demonstrate either poor to moderate selectivity versus the broader human kinome or physicochemical properties unsuitable for use in in vitro systems or in vivo models. A nonconventional analysis of data from a GSK3ß inhibitor high-throughput screening campaign, which excluded known GSK3 inhibitor chemotypes, led to the discovery of a novel pyrazolo-tetrahydroquinolinone scaffold with unparalleled kinome-wide selectivity for the GSK3 kinases. Taking advantage of an uncommon tridentate interaction with the hinge region of GSK3, we developed highly selective and potent GSK3 inhibitors, BRD1652 and BRD0209, which demonstrated in vivo efficacy in a dopaminergic signaling paradigm modeling mood-related disorders. These new chemical probes open the way for exclusive analyses of the function of GSK3 kinases in multiple signaling pathways involved in many prevalent disorders.


Sujet(s)
Glycogen Synthase Kinase 3/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Animaux , Conception de médicament , Humains
19.
ACS Chem Biol ; 11(2): 363-74, 2016 Feb 19.
Article de Anglais | MEDLINE | ID: mdl-26640968

RÉSUMÉ

Modulation of histone deacetylase (HDAC) activity has been implicated as a potential therapeutic strategy for multiple diseases. However, it has been difficult to dissect the role of individual HDACs due to a lack of selective small-molecule inhibitors. Here, we report the synthesis of a series of highly potent and isoform-selective class I HDAC inhibitors, rationally designed by exploiting minimal structural changes to the clinically experienced HDAC inhibitor CI-994. We used this toolkit of isochemogenic or chemically matched inhibitors to probe the role of class I HDACs in ß-cell pathobiology and demonstrate for the first time that selective inhibition of an individual HDAC isoform retains beneficial biological activity and mitigates mechanism-based toxicities. The highly selective HDAC3 inhibitor BRD3308 suppressed pancreatic ß-cell apoptosis induced by inflammatory cytokines, as expected, or now glucolipotoxic stress, and increased functional insulin release. In addition, BRD3308 had no effect on human megakaryocyte differentiation, while inhibitors of HDAC1 and 2 were toxic. Our findings demonstrate that the selective inhibition of HDAC3 represents a potential path forward as a therapy to protect pancreatic ß-cells from inflammatory cytokines and nutrient overload in diabetes.


Sujet(s)
Cytoprotection/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/pharmacologie , Cellules à insuline/effets des médicaments et des substances chimiques , Séquence d'acides aminés , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Conception de médicament , Inhibiteurs de désacétylase d'histone/pharmacocinétique , Histone deacetylases/composition chimique , Histone deacetylases/métabolisme , Humains , Cellules à insuline/cytologie , Données de séquences moléculaires , Isoformes de protéines/composition chimique , Isoformes de protéines/métabolisme , Rats
20.
Blood ; 126(16): 1930-9, 2015 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-26320100

RÉSUMÉ

Fetal hemoglobin (HbF, α2γ2) induction is a well-validated strategy for sickle cell disease (SCD) treatment. Using a small-molecule screen, we found that UNC0638, a selective inhibitor of EHMT1 and EHMT2 histone methyltransferases, induces γ-globin expression. EHMT1/2 catalyze mono- and dimethylation of lysine 9 on histone 3 (H3K9), raising the possibility that H3K9Me2, a repressive chromatin mark, plays a role in silencing γ-globin expression. In primary human adult erythroid cells, UNC0638 and EHMT1 or EHMT2 short hairpin RNA-mediated knockdown significantly increased γ-globin expression, HbF synthesis, and the percentage of cells expressing HbF. At effective concentrations, UNC0638 did not alter cell morphology, proliferation, or erythroid differentiation of primary human CD34(+) hematopoietic stem and progenitor cells in culture ex vivo. In murine erythroleukemia cells, UNC0638 and Ehmt2 CRISPR/Cas9-mediated knockout both led to a marked increase in expression of embryonic ß-globin genes Hbb-εy and Hbb-ßh1. In primary human adult erythroblasts, chromatin immunoprecipitation followed by sequencing analysis revealed that UNC0638 treatment leads to genome-wide depletion in H3K9Me2 and a concomitant increase in the activating mark H3K9Ac, which was especially pronounced at the γ-globin gene region. In RNA-sequencing analysis of erythroblasts, γ-globin genes were among the most significantly upregulated genes by UNC0638. Further increase in γ-globin expression in primary human adult erythroid cells was achieved by combining EHMT1/2 inhibition with the histone deacetylase inhibitor entinostat or hypomethylating agent decitabine. Our data provide genetic and pharmacologic evidence that EHMT1 and EHMT2 are epigenetic regulators involved in γ-globin repression and represent a novel therapeutic target for SCD.


Sujet(s)
Épigenèse génétique/effets des médicaments et des substances chimiques , Érythroblastes/métabolisme , Hémoglobine foetale/biosynthèse , Histone-lysine N-methyltransferase/antagonistes et inhibiteurs , Quinazolines/pharmacologie , Drépanocytose/traitement médicamenteux , Drépanocytose/métabolisme , Animaux , Lignée cellulaire tumorale , Érythroblastes/cytologie , Cellules érythroïdes/cytologie , Cellules érythroïdes/métabolisme , Femelle , Antigènes d'histocompatibilité/métabolisme , Histone-lysine N-methyltransferase/métabolisme , Humains , Mâle , Souris
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...